1
|
Han NN, Wang XP, Jin JA, Li WH, Yang WY, Fan NS, Jin RC. Underrated risk of antibiotic resistance genes dissemination mediated by bioaerosols released from anaerobic biological wastewater treatment system. WATER RESEARCH 2025; 279:123463. [PMID: 40073489 DOI: 10.1016/j.watres.2025.123463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Antibiotic resistance has been recognized as one of the most prevalent public health problems. The bioaerosol-mediated spread of antibiotic resistance genes (ARGs) is an important but underrated pathway. Therefore, this work investigated the comprehensive resistome and pathogen-induced risk in bioaerosols released from anaerobic ammonium oxidation (anammox) process under antibiotic stress. The results showed that the bioaerosol oxidation potential increased by 2.7 times after the addition of sulfamethoxazole (SMX) into the anammox system. Based on the metagenomic analyses, abundant ARGs were enriched in bioaerosols, especially novA, olec, msbA and patA. There were many antibiotic resistance contigs carrying at least two mobile genetic elements (MGEs) in bioaerosols. Compared to the control, SMX caused the significant increase in ARGs proportion in plasmids from 11.4 % to 19.4 %. Similarly, the abundance of the type IV secretion system protein encoding genes (mtrA and mtrB) increased by 30.2 % and 31.5 %, respectively, which was conducive to gene transfer between bacteria. In addition, SMX stress induced the reactive oxygen species (ROS) production and the upregulation of genes related to membrane protein and DNA replication, further facilitating ARGs transfer. The co-occurrence networks showed that Aquamicrobium and Microbacterium probably were the hosts of most ARGs. Notably, four abundant human pathogens were detected in bioaerosols from the anammox system, which raised concerns on the health risk of resistant bioaerosol diffusion. These findings reveal the potential of horizontal gene transfer through bioaerosols and provide a guidance for systematically assessing the risk of environmental antibiotic resistance and relevant pathogens.
Collapse
Affiliation(s)
- Na-Na Han
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Xue-Ping Wang
- School of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Jing-Ao Jin
- School of Engineering, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Wen-Hui Li
- School of Engineering, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Wen-Ya Yang
- School of Engineering, Hangzhou Normal University, Hangzhou 311121, PR China
| | - Nian-Si Fan
- School of Engineering, Hangzhou Normal University, Hangzhou 311121, PR China.
| | - Ren-Cun Jin
- School of Engineering, Hangzhou Normal University, Hangzhou 311121, PR China
| |
Collapse
|
2
|
Yang F, Li M, Wu H, Yu C, Liu W, Chen H. Comparative genomics-based insights into Pantoea ananatis strains, isolated from white spot diseased leaves of maize with plant growth-promoting attributes. Appl Environ Microbiol 2025:e0032925. [PMID: 40387325 DOI: 10.1128/aem.00329-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/13/2025] [Indexed: 05/20/2025] Open
Abstract
Pantoea ananatis is a member of the Enterobacteriaceae family known for its broad host adaptability. This study isolated 10 P. ananatis strains from white spot (MWS)-diseased leaves of maize (Zea mays) grown in Yunnan Province, China, and analyzed their putative functions, genomic diversity, and variation. The inoculation tests revealed that none of the 10 isolates caused MWS symptoms in maize. Nine maize isolates, except for S47, induced a hypersensitive response (HR) in tobacco and caused rot symptoms in onion. Most isolates exhibited plant growth-promoting characteristics, with strains JCC14, JCY1, and S47 significantly enhancing maize seedling growth parameters. Genomic sequencing of 10 maize isolates and two rice isolates revealed that 12 isolates clustered into three groups, with an open pan-genome identified. Ancestral reconstruction indicated that the genome size increased in Group A and then decreased in Group B, with significant gains in orthologous groups at Node 14, the most recent common ancestor (MRCA) of Group A and Group B, and at Node 19, the MRCA of seven maize-isolated strains and other Group B strains. Additionally, 11 single-copy orthologous groups were under positive selection. Furthermore, the HIVir (high virulence, also known as PASVIL, P. ananatis-specific virulence locus) cluster and type VI secretion system-related genes were conserved in certain P. ananatis strains but were not related to their group divergences. This study not only reveals the diverse functions of MWS-diseased maize P. ananatis isolates, but also enhances our understanding of divergent genome evolution and environmental adaptation across P. ananatis species.IMPORTANCEPantoea ananatis is a bacterium commonly found in various agronomic crops. Maize white spot (MWS) has been one of the most destructive diseases affecting maize, leading to significant economic losses. This study clarified that P. ananatis strains colonized maize leaves but were not the causal agents of MWS in Yunnan Province, China. Moreover, most of these P. ananatis strains exhibited plant growth-promoting (PGP) activities, induced hypersensitive response (HR) activity on tobacco, and caused rot symptoms in onion. Notably, the analysis of divergence throughout the evolutionary process revealed significant genomic evolution and environmental adaptation in these P. ananatis strains. This highlights the genetic exchange that has shaped the genome of P. ananatis. These findings improve our understanding of the functional diversity of P. ananatis strains across different hosts and their positions within the evolutionary lineages of P. ananatis species.
Collapse
Affiliation(s)
- Fenghuan Yang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Miao Li
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hanxiang Wu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Chao Yu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Wende Liu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Huamin Chen
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
3
|
Trepka KR, Kidder WA, Kyaw TS, Halsey T, Olson CA, Ortega EF, Noecker C, Upadhyay V, Stanfield D, Steiding P, Guthrie BGH, Spanogiannopoulos P, Dumlao D, Turnbaugh JA, Stachler MD, Van Blarigan EL, Venook AP, Atreya CE, Turnbaugh PJ. Expansion of a bacterial operon during cancer treatment ameliorates fluoropyrimidine toxicity. Sci Transl Med 2025; 17:eadq8870. [PMID: 40238917 DOI: 10.1126/scitranslmed.adq8870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/15/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025]
Abstract
Dose-limiting toxicities remain a major barrier to drug development and therapy, revealing the limited predictive power of human genetics. Here, we demonstrate the utility of a more comprehensive approach to studying drug toxicity through longitudinal profiling of the human gut microbiome during colorectal cancer (CRC) treatment (NCT04054908) coupled to cell culture and mouse experiments. Substantial shifts in gut microbial community structure during oral fluoropyrimidine treatment across multiple patient cohorts, in mouse small and large intestinal contents, and in patient-derived ex vivo communities were revealed by 16S rRNA gene sequencing. Metagenomic sequencing revealed marked shifts in pyrimidine-related gene abundance during oral fluoropyrimidine treatment, including enrichment of the preTA operon, which was sufficient for the inactivation of active metabolite 5-fluorouracil (5-FU). preTA+ bacteria depleted 5-FU in gut microbiota grown ex vivo and in the mouse distal gut. Germ-free and antibiotic-treated mice experienced increased fluoropyrimidine toxicity, which was rescued by colonization with the mouse gut microbiota, preTA+ Escherichia coli, or preTA-high stool from patients with CRC. Last, preTA abundance was negatively associated with fluoropyrimidine toxicity in patients. Together, these data support a causal, clinically relevant interaction between a human gut bacterial operon and the dose-limiting side effects of cancer treatment. Our approach may be generalizable to other drugs, including cancer immunotherapies, and provides valuable insights into host-microbiome interactions in the context of disease.
Collapse
Affiliation(s)
- Kai R Trepka
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Wesley A Kidder
- Department of Medicine, Division of Hematology and Oncology, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA
| | - Than S Kyaw
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Taylor Halsey
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christine A Olson
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Edwin F Ortega
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Cecilia Noecker
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Vaibhav Upadhyay
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dalila Stanfield
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA
| | - Paige Steiding
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA
| | - Benjamin G H Guthrie
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Peter Spanogiannopoulos
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Darren Dumlao
- Department of Gastroenterology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jessie A Turnbaugh
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew D Stachler
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Erin L Van Blarigan
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alan P Venook
- Department of Medicine, Division of Hematology and Oncology, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA
| | - Chloe E Atreya
- Department of Medicine, Division of Hematology and Oncology, University of California, San Francisco, San Francisco, CA 94143, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA
| | - Peter J Turnbaugh
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
4
|
Lin SL, Nie QC, Law COK, Pham HQ, Chau HF, Lau TCK. A novel plasmid-encoded transposon-derived small RNA reveals the mechanism of sRNA-regulated bacterial persistence. mBio 2025; 16:e0381424. [PMID: 39998215 PMCID: PMC11980398 DOI: 10.1128/mbio.03814-24] [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: 12/11/2024] [Accepted: 02/03/2025] [Indexed: 02/26/2025] Open
Abstract
Small regulatory RNAs (sRNAs) in bacteria are crucial for controlling various cellular functions and provide immediate response to the environmental stresses. Antibiotic persistence is a phenomenon that a small subpopulation of bacteria survives under the exposure of a lethal concentration of antibiotics, potentially leading to the development of drug resistance in bacteria. Here, we reported a novel transposon-derived sRNA called stnpA, which can modulate fosfomycin persistence of the bacteria. The stnpA sRNA located in the transposon with its own promoter is highly conserved among the prevalent multidrug resistance (MDR) plasmids in various pathogenic bacteria and expressed in response to the fosfomycin stress. It can directly bind to the ABC transporter, YadG, whereas this protein-RNA interaction modulated the export of fosfomycin and led to the enhancement of bacterial persistence. According to our knowledge, stnpA is the first identified transposon-derived sRNA, which controlled antibiotic persistence of bacteria, and our work demonstrated that nonresistance genes on MDR plasmids such as plasmid-encoded sRNA can provide additional survival advantages to the bacterial host against the antibiotics. In addition, the stnpA sRNA can be potentially utilized as the druggable target for the development of novel therapeutic strategies to overcome bacterial persistence. IMPORTANCE This study unveils a groundbreaking discovery in the realm of bacterial antibiotic persistence, highlighting the pivotal role of a newly identified small RNA (sRNA) called stnpA, which is a multidrug resistance plasmid-encoded transposon-derived sRNA that interacts directly with ABC transporter YadG to modulate the efflux of fosfomycin. Our findings elucidate a novel mechanism of small RNA-regulated fosfomycin persistence in bacteria that provides the potential pathway for the emergence of drug resistance in bacteria upon antibiotic treatment. Importantly, this study provides the first example of linking sRNA regulation to antibiotic persistence, presenting stnpA sRNA as a potential therapeutic target. This study underscores the critical role of noncoding RNAs in bacterial adaptation and offers valuable insights for developing new strategies to combat antibiotic persistence.
Collapse
Affiliation(s)
- Shu-Ling Lin
- Department of Biomedical Sciences, College of Biomedicine, City University of Hong Kong, Hong Kong, China
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China
| | - Qi-Chang Nie
- Department of Biomedical Sciences, College of Biomedicine, City University of Hong Kong, Hong Kong, China
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China
| | - Carmen Oi-Kwan Law
- Department of Biomedical Sciences, College of Biomedicine, City University of Hong Kong, Hong Kong, China
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China
| | - Hoa-Quynh Pham
- Department of Biomedical Sciences, College of Biomedicine, City University of Hong Kong, Hong Kong, China
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China
| | - Ho-Fai Chau
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Terrence Chi-Kong Lau
- Department of Biomedical Sciences, College of Biomedicine, City University of Hong Kong, Hong Kong, China
- Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
5
|
Yun H, Seo JH, Kim YK, Yang J. Examining the bacterial diversity including extracellular vesicles in air and soil: implications for human health. PLoS One 2025; 20:e0320916. [PMID: 40168325 PMCID: PMC11960916 DOI: 10.1371/journal.pone.0320916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/26/2025] [Indexed: 04/03/2025] Open
Abstract
As the significance of human health continues to rise, the microbiome has shifted its focus from microbial composition to the functional roles it plays. In parallel, interest in ultrafine particles associated with clinically important impact has been increasing. Bacterial extracellular vesicles (BEVs), involved in systemic microbiome activity, are nano-sized spherical vesicles (20 - 100 nm in diameter) containing DNA, RNA, proteins, and lipids. They are known to be absorbed into the body potentially through air and soil, circulate in the blood, and directly impact diseases by affecting organs. Therefore, the aim of this study is to examine the biodiversity of bacteria and BEVs and predicted functional pathways. We sampled air and soil samples in Seoul, Korea and analyzed metagenomics based on 16S rRNA sequencing. At the phylum levels, Firmicutes in BEVs from soil and air were significantly higher than in bacteria, and Acidobacteria in both bacteria and BEVs from soil were significantly higher than from air (p < 0.05). The most dominant genera were Pseudomonas in bacteria from air and soil; and Escherichia-Shigella in BEVs from air and soil. In addition, Two-component system (ko02020) and ATP-binding cassette transporters (ko02010) were dominant functional pathways in both air and soil. The most functional pathways and orthologous groups were significantly different between air and soil (p < 0.05). In conclusion, human health can be affected differently depending on type of environment. Future study is necessary to have a better understanding of human health effects from environmental microbiota.
Collapse
Affiliation(s)
- Hyunjun Yun
- The AI Convergence Appliances Research Center, Korea Electronics Technology Institute, Gwangju, Republic of Korea
| | - Ji Hoon Seo
- Department of Environmental Health, Korea University, Seoul, Republic of Korea
| | | | - Jinho Yang
- Department of Occupational Health and Safety, Semyung University, Jecheon-si, Chungcheongbuk-do, Republic of Korea
| |
Collapse
|
6
|
Meyer AC, McIlvin MR, Lopez P, Searle BC, Saito MA. Proteomic profiling of zinc homeostasis mechanisms in Pseudomonas aeruginosa through data-dependent and data-independent acquisition mass spectrometry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632865. [PMID: 39868216 PMCID: PMC11761036 DOI: 10.1101/2025.01.13.632865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Zinc is central to the function of many proteins, yet the mechanisms of zinc homeostasis and their interplay with other cellular systems remain underexplored. In this study, we employ data-dependent acquisition (DDA) and data-independent acquisition (DIA) mass spectrometry to investigate proteome changes in Pseudomonas aeruginosa under conditions of different zinc availability. Using these methods, we detected 2143 unique proteins, 1578 of which were identified by both DDA and DIA. We demonstrated that most of the previously described Zn homeostasis systems exhibit proteomic responses that follow similar trends to those seen in transcriptomics studies. However, some proteins that are considered instrumental in Zn homeostasis, notably those in Zn transporter ZnuABC, were not detected by our methods, although other proteins of other uptake systems were abundant. Furthermore, changes in abundance of multiple Zn-metalloproteins and Zn-independent homologs were clearly observable, with respective increases and decreases when Zn was provided, though the magnitude of these changes varied. Most of the Zn-metalloproteins observed were located in one of two Zur-regulated operons between PA5534 and PA5541. This study provides a view of Zn homeostasis mechanisms that is complementary to existing transcriptomics investigations: as gene transcripts are not strictly proportional to the actual distribution of proteins within a cell, analysis of the proteome offers another way to assess the relative use and importance of similar or ostensibly redundant systems in different conditions and can highlight shifts in metal prioritization between metalloproteins.
Collapse
Affiliation(s)
- Annaliese Cs Meyer
- Woods Hole Oceanographic Institution, Department of Marine Chemistry and Geochemistry, Woods Hole, MA
- Massachusetts Institute of Technology, Department of Earth, Atmospheric, and Planetary Sciences, Cambridge, MA
- The MIT-WHOI Joint Program in Oceanography/Applied Ocean Science and Engineering, Woods Hole, MA
| | - Matthew R McIlvin
- Woods Hole Oceanographic Institution, Department of Marine Chemistry and Geochemistry, Woods Hole, MA
| | - Paloma Lopez
- Woods Hole Oceanographic Institution, Department of Marine Chemistry and Geochemistry, Woods Hole, MA
| | - Brian C Searle
- Ohio State University Medical Center, Department of Biomedical Informatics, Columbus, OH
- Ohio State University, Department of Chemistry and Biochemistry, Columbus, OH
| | - Mak A Saito
- Woods Hole Oceanographic Institution, Department of Marine Chemistry and Geochemistry, Woods Hole, MA
| |
Collapse
|
7
|
Lavergne M, Schaerer R, De Grandis S, Bouheraoua S, Adenuga O, Muralt T, Schaerer T, Chèvre L, Failla A, Matthey P, Stumpe M, Kressler D, Mantel PY, Walch M. Executioner caspases degrade essential mediators of pathogen-host interactions to inhibit growth of intracellular Listeria monocytogenes. Cell Death Dis 2025; 16:55. [PMID: 39885151 PMCID: PMC11782612 DOI: 10.1038/s41419-025-07365-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/20/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025]
Abstract
Cell death mediated by executioner caspases is essential during organ development and for organismal homeostasis. The mechanistic role of activated executioner caspases in antibacterial defense during infections with intracellular bacteria, such as Listeria monocytogenes, remains elusive. Cell death upon intracellular bacterial infections is considered altruistic to deprive the pathogens of their protective niche. To establish infections in a human host, Listeria monocytogenes deploy virulence mediators, including membranolytic listeriolysin O (LLO) and the invasion associated protein p60 (Iap), allowing phagosomal escape, intracellular replication and cell-to-cell spread. Here, by means of chemical and genetical modifications, we show that the executioner caspases-3 and -7 efficiently inhibit growth of intracellular Listeria monocytogenes in host cells. Comprehensive proteomics revealed multiple caspase-3 substrates in the Listeria secretome, including LLO, Iap and various other proteins crucially involved in pathogen-host interactions. Listeria secreting caspase-uncleavable LLO or Iap gained significant growth advantage in epithelial cells. With that, we uncovered an underappreciated defense barrier and a non-canonical role of executioner caspases to degrade virulence mediators, thus impairing intracellular Listeria growth.
Collapse
Affiliation(s)
- Marilyne Lavergne
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Raffael Schaerer
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Sara De Grandis
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Safaa Bouheraoua
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Oluwadamilola Adenuga
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Tanja Muralt
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Tiffany Schaerer
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Léa Chèvre
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Alessandro Failla
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Patricia Matthey
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Michael Stumpe
- Faculty of Science and Medicine, Department of Biology, Metabolomics and Proteomics Platform, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Dieter Kressler
- Faculty of Science and Medicine, Department of Biology, Metabolomics and Proteomics Platform, University of Fribourg, CH-1700, Fribourg, Switzerland
| | - Pierre-Yves Mantel
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland
- Christine Kühne - Center for Allergy Research and Education (CK-CARE), CH-7265, Davos Wolfgang, Switzerland
| | - Michael Walch
- Faculty of Science and Medicine, Department of Oncology, Microbiology and Immunology, Anatomy unit, University of Fribourg, CH-1700, Fribourg, Switzerland.
| |
Collapse
|
8
|
Chen J, Wu Y, Zhu Y, Zhang L, Xu Y, Liu Y. Adaptation for Staphylococcus aureus to hosts via insertion mutation in the accessory gene regulator agrC gene: decreased virulence and enhanced persistence capacity. Microbiol Spectr 2025; 13:e0149724. [PMID: 39611824 PMCID: PMC11705864 DOI: 10.1128/spectrum.01497-24] [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: 06/19/2024] [Accepted: 11/01/2024] [Indexed: 11/30/2024] Open
Abstract
Staphylococcus aureus is an important human pathogen due to its vast array of virulence factors regulated by multiple regulatory mechanisms, including the accessory gene regulator. In this study, two S. aureus strains were simultaneously isolated from the blood of a febrile patient, belonging to the same clone, designated as 23H with a complete hemolytic phenotype, and 23B, exhibiting an incomplete hemolytic phenotype. The genomic comparison between strains 23B and 23H revealed that 23B had a single adenine base insertion at position 923 in the agrC gene, leading to a functional loss of the encoded AgrC. Experimental findings showed that strain 23B had decreased hemolytic activity, lower cytotoxicity against human alveolar epithelial A549 cells and in the Galleria mellonella model, and a reduced ability to survive intracellularly after infecting macrophages, in comparison to 23H. Conversely, 23B exhibited enhanced biofilm formation, greater adherence to A549 cells, and increased persistence in the face of vancomycin and daptomycin treatment. Transcriptomic analysis revealed that 23B upregulated surface protein-encoding genes while simultaneously reducing the expression levels of virulence factors, highlighting the intricate regulatory adjustments facilitating its persistence and reducing pathogenic potential. ATP assay results indicated that 23B maintained elevated ATP levels during the exponential phase yet exhibited reduced levels in the stationary phase when compared with 23H. Our findings suggested that the mutation in the agrC gene of S. aureus results in diminished virulence but markedly enhances persistence. This mutated strain warrants clinical attention because it may lead to treatment failures and persist in patients. IMPORTANCE In clinical antimicrobial therapy, bacterial strains often develop resistance to antimicrobial agents. Additionally, mutations in their gene regulatory networks can increase their persistence, especially in immunocompromised patients. This study identified an insertion mutation in the accessory gene regulator, agrC gene, carried by a Staphylococcus aureus strain isolated from the blood of a febrile patient, leading to the functional loss of AgrC. Further research revealed that despite the reduced virulence of the mutated strain, it significantly bolstered the capacity to adapt and endure within the host during prolonged infections. This was evidenced by increased adhesion and biofilm formation capabilities, development of antimicrobial tolerance, and decreased ATP levels linked to persistence. Therefore, monitoring these mutations in S. aureus is crucial clinically, as they can complicate treatment strategies.
Collapse
Affiliation(s)
- Jiawei Chen
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yun Wu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Zhu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Zhang
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingchun Xu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yali Liu
- Department of Laboratory Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
HOW SS, CHIENG S, NATHAN S, LAM SD. ATP-binding cassette (ABC) transporters: structures and roles in bacterial pathogenesis. J Zhejiang Univ Sci B 2024; 26:58-75. [PMID: 39815611 PMCID: PMC11735909 DOI: 10.1631/jzus.b2300641] [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: 09/12/2023] [Accepted: 12/17/2023] [Indexed: 10/22/2024]
Abstract
Adenosine triphosphate (ATP)-binding cassette (ABC) transporter systems are divided into importers and exporters that facilitate the movement of diverse substrate molecules across the lipid bilayer, against the concentration gradient. These transporters comprise two highly conserved nucleotide-binding domains (NBDs) and two transmembrane domains (TMDs). Unlike ABC exporters, prokaryotic ABC importers require an additional substrate-binding protein (SBP) as a recognition site for specific substrate translocation. The discovery of a large number of ABC systems in bacterial pathogens revealed that these transporters are crucial for the establishment of bacterial infections. The existing literature has highlighted the roles of ABC transporters in bacterial growth, pathogenesis, and virulence. These roles include importing essential nutrients required for a variety of cellular processes and exporting outer membrane-associated virulence factors and antimicrobial substances. This review outlines the general structures and classification of ABC systems to provide a comprehensive view of the activities and roles of ABC transporters associated with bacterial virulence and pathogenesis during infection.
Collapse
|
10
|
Trepka KR, Kidder WA, Kyaw TS, Halsey T, Olson CA, Ortega EF, Noecker C, Upadhyay V, Stanfield D, Steiding P, Guthrie BGH, Spanogiannopoulos P, Dumlao D, Turnbaugh JA, Stachler MD, Van Blarigan EL, Venook AP, Atreya CE, Turnbaugh PJ. Expansion of a bacterial operon during cancer treatment ameliorates drug toxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597471. [PMID: 38895199 PMCID: PMC11185696 DOI: 10.1101/2024.06.04.597471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Dose-limiting toxicities remain a major barrier to drug development and therapy, revealing the limited predictive power of human genetics. Herein, we demonstrate the utility of a more comprehensive approach to studying drug toxicity through longitudinal study of the human gut microbiome during colorectal cancer (CRC) treatment (NCT04054908) coupled to cell culture and mouse experiments. 16S rRNA gene sequencing revealed significant shifts in gut microbial community structure during oral fluoropyrimidine treatment across multiple patient cohorts, in mouse small and large intestinal contents, and in patient-derived ex vivo communities. Metagenomic sequencing revealed marked shifts in pyrimidine-related gene abundance during oral fluoropyrimidine treatment, including enrichment of the preTA operon, which is sufficient for the inactivation of active metabolite 5-fluorouracil (5-FU). preTA + bacteria depleted 5-FU in gut microbiota grown ex vivo and the mouse distal gut. Germ-free and antibiotic-treated mice experienced increased fluoropyrimidine toxicity, which was rescued by colonization with the mouse gut microbiota, preTA + E. coli, or preTA-high CRC patient stool. Finally, preTA abundance was negatively associated with fluoropyrimidine toxicity in patients. Together, these data support a causal, clinically relevant interaction between a human gut bacterial operon and the dose-limiting side effects of cancer treatment. Our approach is generalizable to other drugs, including cancer immunotherapies, and provides valuable insights into host-microbiome interactions in the context of disease.
Collapse
Affiliation(s)
- Kai R. Trepka
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, USA
| | - Wesley A. Kidder
- Department of Medicine, Division of Hematology and Oncology, University of California San Francisco; San Francisco, USA
- UCSF Helen Diller Family Comprehensive Cancer Center; San Francisco, USA
| | - Than S. Kyaw
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, USA
| | - Taylor Halsey
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, USA
| | - Christine A. Olson
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, USA
| | - Edwin F. Ortega
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, USA
| | - Cecilia Noecker
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, USA
| | - Vaibhav Upadhyay
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, USA
| | - Dalila Stanfield
- UCSF Helen Diller Family Comprehensive Cancer Center; San Francisco, USA
| | - Paige Steiding
- UCSF Helen Diller Family Comprehensive Cancer Center; San Francisco, USA
| | - Benjamin G. H. Guthrie
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, USA
| | - Peter Spanogiannopoulos
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, USA
| | - Darren Dumlao
- Department of Gastroenterology, University of California San Francisco; San Francisco, USA
| | - Jessie A. Turnbaugh
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, USA
| | - Matthew D. Stachler
- Department of Pathology, University of California San Francisco; San Francisco, USA
| | - Erin L. Van Blarigan
- UCSF Helen Diller Family Comprehensive Cancer Center; San Francisco, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco; San Francisco, USA
- Department of Urology, University of California San Francisco; San Francisco, USA
| | - Alan P. Venook
- Department of Medicine, Division of Hematology and Oncology, University of California San Francisco; San Francisco, USA
- UCSF Helen Diller Family Comprehensive Cancer Center; San Francisco, USA
| | - Chloe E. Atreya
- Department of Medicine, Division of Hematology and Oncology, University of California San Francisco; San Francisco, USA
- UCSF Helen Diller Family Comprehensive Cancer Center; San Francisco, USA
| | - Peter J. Turnbaugh
- Department of Microbiology and Immunology, University of California San Francisco; San Francisco, USA
- Chan Zuckerberg Biohub-San Francisco; San Francisco, USA
| |
Collapse
|
11
|
Figueroa-Gonzalez PA, Bornemann TLV, Hinzke T, Maaß S, Trautwein-Schult A, Starke J, Moore CJ, Esser SP, Plewka J, Hesse T, Schmidt TC, Schreiber U, Bor B, Becher D, Probst AJ. Metaproteogenomics resolution of a high-CO 2 aquifer community reveals a complex cellular adaptation of groundwater Gracilibacteria to a host-dependent lifestyle. MICROBIOME 2024; 12:194. [PMID: 39369255 PMCID: PMC11452946 DOI: 10.1186/s40168-024-01889-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 07/29/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Bacteria of the candidate phyla radiation (CPR), constituting about 25% of the bacterial biodiversity, are characterized by small cell size and patchy genomes without complete key metabolic pathways, suggesting a symbiotic lifestyle. Gracilibacteria (BD1-5), which are part of the CPR branch, possess alternate coded genomes and have not yet been cultivated. The lifestyle of Gracilibacteria, their temporal dynamics, and activity in natural ecosystems, particularly in groundwater, has remained largely unexplored. Here, we aimed to investigate Gracilibacteria activity in situ and to discern their lifestyle based on expressed genes, using the metaproteogenome of Gracilibacteria as a function of time in the cold-water geyser Wallender Born in the Volcanic Eifel region in Germany. RESULTS We coupled genome-resolved metagenomics and metaproteomics to investigate a cold-water geyser microbial community enriched in Gracilibacteria across a 12-day time-series. Groundwater was collected and sequentially filtered to fraction CPR and other bacteria. Based on 725 Gbps of metagenomic data, 1129 different ribosomal protein S3 marker genes, and 751 high-quality genomes (123 population genomes after dereplication), we identified dominant bacteria belonging to Gallionellales and Gracilibacteria along with keystone microbes, which were low in genomic abundance but substantially contributing to proteomic abundance. Seven high-quality Gracilibacteria genomes showed typical limitations, such as limited amino acid or nucleotide synthesis, in their central metabolism but no co-occurrence with potential hosts. The genomes of these Gracilibacteria were encoded for a high number of proteins involved in cell to cell interaction, supporting the previously surmised host-dependent lifestyle, e.g., type IV and type II secretion system subunits, transporters, and features related to cell motility, which were also detected on protein level. CONCLUSIONS We here identified microbial keystone taxa in a high-CO2 aquifer, and revealed microbial dynamics of Gracilibacteria. Although Gracilibacteria in this ecosystem did not appear to target specific organisms in this ecosystem due to lack of co-occurrence despite enrichment on 0.2-µm filter fraction, we provide proteomic evidence for the complex machinery behind the host-dependent lifestyle of groundwater Gracilibacteria. Video Abstract.
Collapse
Affiliation(s)
- Perla Abigail Figueroa-Gonzalez
- Environmental Metagenomics, Faculty of Chemistry, Research Center One Health of the University Alliance Ruhr, University of Duisburg-Essen, 45151, Essen, Germany
| | - Till L V Bornemann
- Environmental Metagenomics, Faculty of Chemistry, Research Center One Health of the University Alliance Ruhr, University of Duisburg-Essen, 45151, Essen, Germany
- Centre of Water and Environmental Research (ZWU), University of Duisburg-Essen, 45141, Essen, Germany
| | - Tjorven Hinzke
- Microbial Proteomics, Institute of Microbiology, University of Greifswald, 17489, Greifswald, Germany
- Department of Pathogen Evolution, Helmholtz Institute for One Health, 17489, Greifswald, Germany
- Microbial Physiology and Molecular Biology, Institute of Microbiology, University of Greifswald, Greifswald, 17489, Germany
| | - Sandra Maaß
- Microbial Proteomics, Institute of Microbiology, University of Greifswald, 17489, Greifswald, Germany
| | - Anke Trautwein-Schult
- Microbial Proteomics, Institute of Microbiology, University of Greifswald, 17489, Greifswald, Germany
| | - Joern Starke
- Environmental Metagenomics, Faculty of Chemistry, Research Center One Health of the University Alliance Ruhr, University of Duisburg-Essen, 45151, Essen, Germany
| | - Carrie J Moore
- Environmental Metagenomics, Faculty of Chemistry, Research Center One Health of the University Alliance Ruhr, University of Duisburg-Essen, 45151, Essen, Germany
| | - Sarah P Esser
- Environmental Metagenomics, Faculty of Chemistry, Research Center One Health of the University Alliance Ruhr, University of Duisburg-Essen, 45151, Essen, Germany
| | - Julia Plewka
- Environmental Metagenomics, Faculty of Chemistry, Research Center One Health of the University Alliance Ruhr, University of Duisburg-Essen, 45151, Essen, Germany
| | - Tobias Hesse
- Instrumental Analytical Chemistry and Centre for Water and Environmental Research (ZWU), University of Duisburg-Essen, Essen, 45141, Germany
| | - Torsten C Schmidt
- Centre of Water and Environmental Research (ZWU), University of Duisburg-Essen, 45141, Essen, Germany
- Instrumental Analytical Chemistry and Centre for Water and Environmental Research (ZWU), University of Duisburg-Essen, Essen, 45141, Germany
| | - Ulrich Schreiber
- Department of Geology, University of Duisburg-Essen, 45141, Essen, Germany
| | - Batbileg Bor
- Microbiology, The Forsyth Institute, Cambridge, MA, 02142, USA
| | - Dörte Becher
- Microbial Proteomics, Institute of Microbiology, University of Greifswald, 17489, Greifswald, Germany
| | - Alexander J Probst
- Environmental Metagenomics, Faculty of Chemistry, Research Center One Health of the University Alliance Ruhr, University of Duisburg-Essen, 45151, Essen, Germany.
- Centre of Water and Environmental Research (ZWU), University of Duisburg-Essen, 45141, Essen, Germany.
| |
Collapse
|
12
|
Dong Z, Li C, Tian X, Guo X, Li X, Ren W, Chi J, Zhang L, Li F, Zhu Y, Zhang W, Yan M. Characterization Studies on the sugC Gene of Streptococcus suis Serotype 2 in Adhesion, Invasion, and Virulence in Mice. Vet Sci 2024; 11:447. [PMID: 39330826 PMCID: PMC11435659 DOI: 10.3390/vetsci11090447] [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: 08/03/2024] [Revised: 09/08/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
The sugC gene of Streptococcus suis (S. suis) is a coding gene for the ATP-binding transporter-associated protein with strong pathogenicity. In order to reveal the effect of the sugC gene on the virulence of S. suis serotype 2, a wild-type strain of TJS75, isolated from fattening pigs' brain tissue samples, was used as a parent strain, and a knockout sugC gene (ΔsugC) and complementary strain (CΔsugC) were successfully constructed via homologous recombination technology. The biological characteristics of TJS75, ΔsugC and CΔsugC were compared and analyzed through growth curves, biochemical characteristics, hemolysis characteristics, cell infection tests and pathogenicity tests on BALB/c mice. The results of the growth characteristic experiments in vitro showed that the plateau stage growth period of ΔsugC was delayed compared to the TJS75 strain, but there was no difference in the total number of bacteria. The biochemical characteristics and hemolysis ability of ΔsugC in sheep blood had no difference compared with TJS75, but its adhesion and invasion abilities in PK-15 cells were decreased. Knockout of the sugC gene had no impact on the expression levels of adhesion-related genes in TJS75 in real-time PCR analysis. In addition, the LD50 of ΔsugC in BALB/c mice was 1.47 × 108 CFU, seven times higher than that of TJS75 (LD50 = 2.15 × 107 CFU). These results illustrate that the deletion of sugC reduced the virulence of TJS75 to BALB/c mice, but its role in the adhesion and invasion of PK-15 cells in this strain needs to be further explored. In summary, this study provides evidence that the sugC gene is a virulence-related gene in the S. suis serotype 2 strain and plays a crucial role in the adhesion and invasion of S. suis. This study lays a foundation for the further exploration of the potential virulence factors and pathogenesis of S. suis.
Collapse
Affiliation(s)
- Zhimin Dong
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Z.D.); (C.L.); (X.T.); (X.G.); (W.R.); (J.C.); (L.Z.); (F.L.)
- National Data Center of Animal Health, Tianjin 300381, China
| | - Cheng Li
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Z.D.); (C.L.); (X.T.); (X.G.); (W.R.); (J.C.); (L.Z.); (F.L.)
- National Data Center of Animal Health, Tianjin 300381, China
| | - Xiangxue Tian
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Z.D.); (C.L.); (X.T.); (X.G.); (W.R.); (J.C.); (L.Z.); (F.L.)
- National Data Center of Animal Health, Tianjin 300381, China
| | - Xiaoran Guo
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Z.D.); (C.L.); (X.T.); (X.G.); (W.R.); (J.C.); (L.Z.); (F.L.)
- National Data Center of Animal Health, Tianjin 300381, China
| | - Xiuli Li
- Institute of Agro-Product Safety and Nutrition, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China;
| | - Weike Ren
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Z.D.); (C.L.); (X.T.); (X.G.); (W.R.); (J.C.); (L.Z.); (F.L.)
- National Data Center of Animal Health, Tianjin 300381, China
| | - Jingjing Chi
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Z.D.); (C.L.); (X.T.); (X.G.); (W.R.); (J.C.); (L.Z.); (F.L.)
- National Data Center of Animal Health, Tianjin 300381, China
| | - Li Zhang
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Z.D.); (C.L.); (X.T.); (X.G.); (W.R.); (J.C.); (L.Z.); (F.L.)
- National Data Center of Animal Health, Tianjin 300381, China
| | - Fuqiang Li
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Z.D.); (C.L.); (X.T.); (X.G.); (W.R.); (J.C.); (L.Z.); (F.L.)
- National Data Center of Animal Health, Tianjin 300381, China
| | - Yao Zhu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (Y.Z.); (W.Z.)
| | - Wanjiang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; (Y.Z.); (W.Z.)
| | - Minghua Yan
- Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China; (Z.D.); (C.L.); (X.T.); (X.G.); (W.R.); (J.C.); (L.Z.); (F.L.)
- National Data Center of Animal Health, Tianjin 300381, China
| |
Collapse
|
13
|
Kulig K, Kowalik K, Surowiec M, Karnas E, Barczyk-Woznicka O, Zuba-Surma E, Pyza E, Kozik A, Rapala-Kozik M, Karkowska-Kuleta J. Isolation and Characteristics of Extracellular Vesicles Produced by Probiotics: Yeast Saccharomyces boulardii CNCM I-745 and Bacterium Streptococcus salivarius K12. Probiotics Antimicrob Proteins 2024; 16:936-948. [PMID: 37209320 PMCID: PMC11126510 DOI: 10.1007/s12602-023-10085-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2023] [Indexed: 05/22/2023]
Abstract
Numerous probiotic microorganisms have repeatedly been shown to produce nanometer-sized structures named extracellular vesicles (EVs). Recently, it has been suggested that similarly to whole microbial cells, EVs produced by probiotics may also demonstrate health benefits to the host, while their application does not involve the risk of infection caused by live microorganisms. In this work, we isolated EVs from two probiotic species originating from different taxonomic domains - yeast Saccharomyces boulardii CNCM I-745 and bacterium Streptococcus salivarius K12. The diameters of S. boulardii EVs were about 142 nm and for S. salivarius EVs about 123 nm. For S. boulardii EVs, 1641 proteins and for S. salivarius EVs, 466 proteins were identified with a liquid chromatography-coupled tandem mass spectrometry and then functionally classified. In both microbial species, metabolic proteins significantly contributed to the cargo of EVs comprising 25% and 26% of all identified vesicular proteins for fungi and bacteria, respectively. Moreover, enzymes associated with cell wall rearrangement, including enzymatically active glucanases, were also identified in EVs. Furthermore, probiotic EVs were shown to influence host cells and stimulate the production of IL-1β and IL-8 by the human monocytic cell line THP-1, and, at the same time, did not cause any remarkable reduction in the survival rate of Galleria mellonella larvae in this invertebrate model commonly used to evaluate microbial EV toxicity. These observations suggest that the EVs produced by the investigated probiotic microorganisms may be promising structures for future use in pro-health applications.
Collapse
Affiliation(s)
- Kamila Kulig
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Katarzyna Kowalik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Magdalena Surowiec
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Kraków, Poland
| | - Elzbieta Karnas
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Olga Barczyk-Woznicka
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Ewa Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Elzbieta Pyza
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Andrzej Kozik
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Maria Rapala-Kozik
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Justyna Karkowska-Kuleta
- Department of Comparative Biochemistry and Bioanalytics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
14
|
Villa M, Wu J, Hansen S, Pahnke J. Emerging Role of ABC Transporters in Glia Cells in Health and Diseases of the Central Nervous System. Cells 2024; 13:740. [PMID: 38727275 PMCID: PMC11083179 DOI: 10.3390/cells13090740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/15/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
ATP-binding cassette (ABC) transporters play a crucial role for the efflux of a wide range of substrates across different cellular membranes. In the central nervous system (CNS), ABC transporters have recently gathered significant attention due to their pivotal involvement in brain physiology and neurodegenerative disorders, such as Alzheimer's disease (AD). Glial cells are fundamental for normal CNS function and engage with several ABC transporters in different ways. Here, we specifically highlight ABC transporters involved in the maintenance of brain homeostasis and their implications in its metabolic regulation. We also show new aspects related to ABC transporter function found in less recognized diseases, such as Huntington's disease (HD) and experimental autoimmune encephalomyelitis (EAE), as a model for multiple sclerosis (MS). Understanding both their impact on the physiological regulation of the CNS and their roles in brain diseases holds promise for uncovering new therapeutic options. Further investigations and preclinical studies are warranted to elucidate the complex interplay between glial ABC transporters and physiological brain functions, potentially leading to effective therapeutic interventions also for rare CNS disorders.
Collapse
Affiliation(s)
- Maria Villa
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, NO-0372 Oslo, Norway
| | - Jingyun Wu
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, NO-0372 Oslo, Norway
| | - Stefanie Hansen
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, NO-0372 Oslo, Norway
| | - Jens Pahnke
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology (PAT), Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, NO-0372 Oslo, Norway
- Institute of Nutritional Medicine (INUM)/Lübeck Institute of Dermatology (LIED), University of Lübeck (UzL) and University Medical Center Schleswig-Holstein (UKSH), Ratzeburger Allee 160, D-23538 Lübeck, Germany
- Department of Pharmacology, Faculty of Medicine, University of Latvia (LU), Jelgavas iela 3, LV-1004 Rīga, Latvia
- School of Neurobiology, Biochemistry and Biophysics, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University (TAU), Tel Aviv IL-6997801, Israel
| |
Collapse
|
15
|
Muñoz-Hisado V, Ruiz-Blas F, Sobrado JM, Garcia-Lopez E, Martinez-Alonso E, Alcázar A, Cid C. Bacterial molecular machinery in the Martian cryosphere conditions. Front Microbiol 2023; 14:1176582. [PMID: 37840745 PMCID: PMC10569478 DOI: 10.3389/fmicb.2023.1176582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/11/2023] [Indexed: 10/17/2023] Open
Abstract
The exploration of Mars is one of the main objectives of space missions since the red planet is considered to be, or was in the past, potentially habitable. Although the surface of Mars is now dry and arid, abundant research suggests that water covered Mars billions of years ago. Recently, the existence of liquid water in subglacial lakes has been postulated below the South pole of Mars. Until now, experiments have been carried out on the survival of microorganisms in Martian surface conditions, but it remains unknown how their adaptation mechanisms would be in the Martian cryosphere. In this work, two bacterial species (Bacillus subtilis and Curtobacterium flacumfaciens) were subjected to a simulated Martian environment during 24 h using a planetary chamber. Afterward, the molecular machinery of both species was studied to investigate how they had been modified. Proteomes, the entire set of proteins expressed by each bacterium under Earth (named standard) conditions and Martian conditions, were compared using proteomic techniques. To establish this evaluation, both the expression levels of each protein, and the variation in their distribution within the different functional categories were considered. The results showed that these bacterial species followed a different strategy. The Bacillus subtilis resistance approach consisted of improving its stress response, membrane bioenergetics, degradation of biomolecules; and to a lesser extent, increasing its mobility and the formation of biofilms or resistance endospores. On the contrary, enduring strategy of Curtobacterium flacumfaciens comprised of strengthening the cell envelope, trying to protect cells from the extracellular environment. These results are especially important due to their implications for planetary protection, missions to Mars and sample return since contamination by microorganisms would invalidate the results of these investigations.
Collapse
Affiliation(s)
| | - Fátima Ruiz-Blas
- GFZ German Research Centre for Geosciences, Section Geomicrobiology, Telegrafenberg, Potsdam, Germany
| | | | | | - Emma Martinez-Alonso
- Hospital Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Alberto Alcázar
- Hospital Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Cristina Cid
- Centro de Astrobiologia (CAB), CSIC-INTA, Madrid, Spain
| |
Collapse
|
16
|
Rao Tatta E, Paul S, Kumavath R. Transcriptome Analysis revealed the Synergism of Novel Rhodethrin inhibition on Biofilm architecture, Antibiotic Resistance and Quorum sensing inEnterococcus faecalis. Gene 2023; 871:147436. [PMID: 37075926 DOI: 10.1016/j.gene.2023.147436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/31/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023]
Abstract
Enterococcus sp. emerged as an opportunistic nosocomial pathogen with the highest antibiotic resistance and mortality rate. Biofilm is problematic primarily since it is regulated by the global bacterial cell to cell communication mediated by the quorum sensing system. sing system. Thus, potential natural antagonists in a novel drug formulation against biofilm-forming Enterococcus faecalis is critical. We used RNA-Seq to evaluate the effects of the novel molecule rhodethrin with chloramphenicol induced on Enterococcus faecalis and DEGs were identified. In transcriptome sequence analysis, a total of 448 with control Vs rhodethrin, 1591 were in control Vs chloramphenicol, 379 genes were DEGs from control Vs synergies, in rhodethrin with chloramphenicol, 379 genes were differentially expressed, whereas 264 genes were significantly downregulated, indicating that 69.69% ofE. faecaliswas altered. The transcriptional sequence data further expression analysis qRT-PCR, and the results shed that the expression profiles of five significant biofilm formation responsible genes such as, Ace, AtpB, lepA, bopD, and typA, 3 genes involved in quorum sensing are sylA, fsrC and camE, and 4 genes involved in resistance were among including liaX, typA, EfrA, and lepA, were significantly suppressed expressions of the biofilm, quorum sensing, and resistance that are supported by transcriptome analysis.
Collapse
Affiliation(s)
- Eswar Rao Tatta
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periya (PO), Kasaragod, Kerala 671320, India
| | - Sayan Paul
- Department of Biochemistry & Molecular Biology, the University of Texas Medical Branch at Galveston, Galveston, Texas 77555, USA
| | - Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Tejaswini Hills, Periya (PO), Kasaragod, Kerala 671320, India; Department of Biotechnology, School of Life Sciences, Pondicherry University, Puducherry 605014, India.
| |
Collapse
|
17
|
Chaudhari R, Singh K, Kodgire P. Biochemical and molecular mechanisms of antibiotic resistance in Salmonella spp. Res Microbiol 2023; 174:103985. [PMID: 35944794 DOI: 10.1016/j.resmic.2022.103985] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 01/11/2023]
Abstract
Salmonella is a diverse Gram-negative bacterium that represents the major disease burden worldwide. According to WHO, Salmonella is one of the fourth global causes of diarrhoeal disease. Antibiotic resistance is a worldwide health concern, and Salmonella spp. is one of the microorganisms that can evade the toxicity of antimicrobials via antibiotic resistance. This review aims to deliver in-depth knowledge of the molecular mechanisms and the underlying biochemical alterations perceived in antibiotic resistance in Salmonella. This information will help understand and mitigate the impact of antibiotic-resistant bacteria on humans and contribute to the state-of-the-art research developing newer and more potent antibiotics.
Collapse
Affiliation(s)
- Rahul Chaudhari
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology, Indore, Simrol, Khandwa Road, Indore 453552, India
| | - Kanika Singh
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology, Indore, Simrol, Khandwa Road, Indore 453552, India
| | - Prashant Kodgire
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology, Indore, Simrol, Khandwa Road, Indore 453552, India.
| |
Collapse
|
18
|
Ngashangva N, Mukherjee PK, Sharma C, Kalita MC, Sarangthem I. Integrated genomics and proteomics analysis of Paenibacillus peoriae IBSD35 and insights into its antimicrobial characteristics. Sci Rep 2022; 12:18861. [PMID: 36344671 PMCID: PMC9640621 DOI: 10.1038/s41598-022-23613-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Antimicrobial resistance has been developing fast and incurring a loss of human life, and there is a need for new antimicrobial agents. Naturally occurring antimicrobial peptides offer the characteristics to counter AMR because the resistance development is low or no resistance. Antimicrobial peptides from Paenibacillus peoriae IBSD35 cell-free supernatant were salted out and purified using chromatography and characterized with liquid chromatography-tandem-mass spectrometry. The extract has shown a high and broad spectrum of antimicrobial activity. Combining the strain IBSD35 genome sequence with its proteomic data enabled the prediction of biosynthetic gene clusters by connecting the peptide from LC-MS/MS data to the gene that encode. Antimicrobial peptide databases offered a platform for the effective search, prediction, and design of AMPs and expanded the studies on their isolation, structure elucidation, biological evaluation, and pathway engineering. The genome-based taxonomy and comparisons have shown that P. peoriae IBSD35 is closely related to Paenibacillus peoriae FSL J3-0120. P. peoriae IBSD35 harbored endophytic trait genes and nonribosomal peptide synthases biosynthetic gene clusters. The comparative genomics revealed evolutionary insights and facilitated the discovery of novel SMs using proteomics from the extract of P. peoriae IBSD35. It will increase the potential to find novel bio-molecules to counter AMR.
Collapse
Affiliation(s)
- Ng Ngashangva
- grid.464584.f0000 0004 0640 0101A National Institute of Department of Biotechnology, Institute of Bioresources and Sustainable Development (IBSD), Govt. of India, Takyelpat, Imphal, Manipur 795001 India
| | - Pulok K. Mukherjee
- grid.464584.f0000 0004 0640 0101A National Institute of Department of Biotechnology, Institute of Bioresources and Sustainable Development (IBSD), Govt. of India, Takyelpat, Imphal, Manipur 795001 India
| | - Chandradev Sharma
- grid.464584.f0000 0004 0640 0101A National Institute of Department of Biotechnology, Institute of Bioresources and Sustainable Development (IBSD), Govt. of India, Takyelpat, Imphal, Manipur 795001 India
| | - Mohan C. Kalita
- grid.411779.d0000 0001 2109 4622Department of Biotechnology, Gauhati University, Jalukbari, Guwahati, Assam 781014 India
| | - Indira Sarangthem
- grid.464584.f0000 0004 0640 0101A National Institute of Department of Biotechnology, Institute of Bioresources and Sustainable Development (IBSD), Govt. of India, Takyelpat, Imphal, Manipur 795001 India
| |
Collapse
|
19
|
Abriouel H, Manetsberger J, Caballero Gómez N, Benomar N. In silico genomic analysis of the potential probiotic Lactiplantibacillus pentosus CF2-10N reveals promising beneficial effects with health promoting properties. Front Microbiol 2022; 13:989824. [PMID: 36406402 PMCID: PMC9670130 DOI: 10.3389/fmicb.2022.989824] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/04/2022] [Indexed: 10/29/2023] Open
Abstract
Lactiplantibacillus pentosus CF2-10 N, isolated from brines of naturally fermented Aloreña green table olives, exhibited high probiotic potential. High throughput sequencing and annotation of genome sequences underline the potential of L. pentosus CF2-10 N as excellent probiotic candidate of vegetable origin. In a previous study we could show the probiotic potential of CF2-10 N in vitro, while in this study in silico analysis of its genome revealed new insights into its safety and functionality. Our findings highlight the microorganism's ecological flexibility and adaptability to a broad range of environmental niches, food matrices and the gastrointestinal tract. These features are shared by both phylogenetically very close L. pentosus strains (CF2-10 N and MP-10) isolated from the same ecological niche with respect to their genome size (≅ 3.6 Mbp), the presence of plasmids (4-5) and several other properties. Nonetheless, additional and unique features are reported in the present study for L. pentosus CF2-10 N. Notably, the safety of L. pentosus CF2-10 N was shown by the absence of virulence determinants and the determination of acquired antibiotic resistance genes, i.e., resistome, which is mostly represented by efflux-pump resistance genes responsible for the intrinsic resistance. On the other hand, defense mechanisms of L. pentosus CF2-10 N include eight prophage regions and a CRISPR/cas system (CRISPR-I and CRISPR-II) as acquired immune system against mobile elements. Finally, the probiotic potential of this strain was further demonstrated by the presence of genes coding for proteins involved in adhesion, exopolysaccharide biosynthesis, tolerance to low pH and bile salts, immunomodulation, and vitamin and enzyme production. Taken together these results, we propose the use of L. pentosus CF2-10 N as a potential and promising probiotic candidate able to colonize several niches and adapt to different lifestyles. The strain can provide attractive functional and probiotic features necessary for its application as starter culture and probiotic.
Collapse
Affiliation(s)
- Hikmate Abriouel
- Área de Microbiología, Departamento de Ciencias de la Salud, Facultad de Ciencias Experimentales, Universidad de Jaén, Jaén, Spain
| | | | | | | |
Collapse
|
20
|
Kerdsin A, Takeuchi D, Akeda Y, Nakamura S, Gottschalk M, Oishi K. Genomic differences between sequence types 1 and 104 of Streptococcus suis Serotype 2. PeerJ 2022; 10:e14144. [PMID: 36221266 PMCID: PMC9548313 DOI: 10.7717/peerj.14144] [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: 01/17/2022] [Accepted: 09/07/2022] [Indexed: 01/21/2023] Open
Abstract
Background Streptococcus suis is a zoonotic pathogen that can cause invasive infections in humans who are in close contact with infected pigs or contaminated pork-derived products. S. suis serotype 2 sequence type (ST) 1 strains are mostly associated with meningitis, whereas ST104 strains are mostly recovered from sepsis cases in humans. No data are available for comparison of the ST1 and ST104 strains at the genomic level, particularly concerning virulence-associated genes. Thus, genomic comparison of both STs was performed in this study. Methods An ST1 isolate (ID26154) from the cerebrospinal fluid of a patient with meningitis and an ST104 isolate (ID24525) from the blood of a patient with sepsis were subjected to shotgun pyrosequencing using the 454 GS Junior System. Genomic comparison was conducted between the ST1 isolate and the ST104 isolate using the Artemis Comparison Tool (ACT) to identify the region of differences (RDs) between ST1 and ST104. Results Fifty-eight RDs were unique to the ST104 genome and were mainly involved in metabolism and cell functional activities, cell wall anchored proteins, bacteriophages and mobile genetic elements, ABC-type transporters, two-component signal transductions, and lantibiotic proteins. Some virulence genes mostly found in ST1 strains were also present in the ST104 genome. Whole-genome comparison is a powerful tool for identifying genomic region differences between different STs of S. suis serotype 2, leading to the identification of the molecular basis of virulence involved in the pathogenesis of the infection.
Collapse
Affiliation(s)
- Anusak Kerdsin
- Faculty of Public Health, Kasetsart University Chalermphrakiat Sakon Nakhon Province Campus, Sakon Nakhon, Thailand
| | - Dan Takeuchi
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yukihiro Akeda
- Research Institute for Microbial Diseases, Osaka University, Osaka, Japan,Division of Infection Control and Prevention, Graduate School of Medicine, Osaka University, Osaka, Japan,Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shota Nakamura
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | | | | |
Collapse
|
21
|
Emergence and spread of antibiotic-resistant foodborne pathogens from farm to table. Food Sci Biotechnol 2022; 31:1481-1499. [PMID: 36065433 PMCID: PMC9435411 DOI: 10.1007/s10068-022-01157-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/26/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Antibiotics have been overused and misused for preventive and therapeutic purposes. Specifically, antibiotics are frequently used as growth promoters for improving productivity and performance of food-producing animals such as pigs, cattle, and poultry. The increasing use of antibiotics has been of great concern worldwide due to the emergence of antibiotic resistant bacteria. Food-producing animals are considered reservoirs for antibiotic resistance genes (ARGs) and residual antibiotics that transfer from the farm through the table. The accumulation of residual antibiotics can lead to additional antibiotic resistance in bacteria. Therefore, this review evaluates the risk of carriage and spread of antibiotic resistance through food chain and the potential impact of antibiotic use in food-producing animals on food safety. This review also includes in-depth discussion of promising antibiotic alternatives such as vaccines, immune modulators, phytochemicals, antimicrobial peptides, probiotics, and bacteriophages.
Collapse
|
22
|
Akhtar AA, Turner DP. The role of bacterial ATP-binding cassette (ABC) transporters in pathogenesis and virulence: Therapeutic and vaccine potential. Microb Pathog 2022; 171:105734. [PMID: 36007845 DOI: 10.1016/j.micpath.2022.105734] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/24/2022]
Abstract
The ATP-binding cassette (ABC) transporter superfamily is found in all domains of life, facilitating critical biological processes through the translocation of a wide variety of substrates from, ions to proteins, across cellular membranes in an ATP-coupled process. The role of ABC transporters in eukaryotes has been well established: the facilitation of genetic diseases and multi-drug resistance (MDR) in cancer patients. In contrast, the role of ABC transporters in prokaryotes has been ambiguous due to their diverse functions and the sheer number of organisms in which they reside. This review examines the role of bacterial ABC transporters in pathogenesis and virulence, and their potential for therapeutic and vaccine application. We demonstrate how ABC transporters play a vital role in the virulence and pathogenesis of several pathogenic bacteria through the import of essential molecules, such as metal ions, amino acids, peptides, vitamins and osmoprotectants, as well as, the export of virulent determinants involved in glycoconjugate biosynthesis and Type I secretion. Furthermore, ABC exporters facilitate the persistence of pathogenic bacteria through the export of toxic xenobiotic substances, thus, contributing to the development of antimicrobial resistance. We also show that ABC transporters display considerable potential for therapeutic application through immunisation and resistance reversal. In conclusion, bacterial ABC transporters play an immense role in virulence and pathogenesis and display desirable traits for clinical use, therefore, potentially aiding in the battle against MDR.
Collapse
Affiliation(s)
- Armaan A Akhtar
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, United Kingdom.
| | - David Pj Turner
- School of Life Sciences, University of Nottingham, University Park, Nottingham, NG7 2RD, United Kingdom
| |
Collapse
|
23
|
Liu Y, Yoo BB, Hwang CA, Martinez MR, Datta AR, Fratamico PM. Involvement of a putative ATP-Binding Cassette (ABC) Involved in manganese transport in virulence of Listeria monocytogenes. PLoS One 2022; 17:e0268924. [PMID: 35617277 PMCID: PMC9135185 DOI: 10.1371/journal.pone.0268924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/10/2022] [Indexed: 11/18/2022] Open
Abstract
Listeria monocytogenes is a foodborne pathogen and the causative agent of listeriosis, a disease associated with high fatality (20–30%) and hospitalization rates (>95%). ATP-Binding Cassette (ABC) transporters have been demonstrated to be involved in the general stress response. In previous studies, in-frame deletion mutants of the ABC transporter genes, LMOf2365_1875 and LMOf2365_1877, were constructed and analyzed; however, additional work is needed to investigate the virulence potential of these deletion mutants. In this study, two in vitro methods and one in vivo model were used to investigate the virulence potential of in-frame deletion mutants of ABC transporter genes. First, the invasion efficiency in host cells was measured using the HT-29 human cell line. Second, cell-to-cell spread activity was measured using a plaque forming assay. Lastly, virulence potential of the mutants was tested in the Galleria mellonella wax moth model. Our results demonstrated that the deletion mutant, ⊿LMOf2365_1875, displayed decreased invasion and cell-to-cell spread efficiency in comparison to the wild-type, LMOf2365, indicating that LMOf2365_1875 may be required for virulence. Furthermore, the reduced virulence of these mutants was confirmed using the Galleria mellonella wax moth model. In addition, the expression levels of 15 virulence and stress-related genes were analyzed by RT-PCR assays using stationary phase cells. Our results showed that virulence-related gene expression levels from the deletion mutants were elevated (15/15 genes from ⊿LMOf2365_1877 and 7/15 genes from ⊿LMOf2365_1875) compared to the wild type LMOf2365, suggesting that ABC transporters may negatively regulate virulence gene expression under specific conditions. The expression level of the stress-related gene, clpE, also was increased in both deletion mutants, indicating the involvement of ABC transporters in the stress response. Taken together, our findings suggest that ABC transporters may be used as potential targets to develop new therapeutic strategies to control L. monocytogenes.
Collapse
Affiliation(s)
- Yanhong Liu
- Eastern Regional Research Center, Agricultural Research Service, U.S. Department of Agriculture, Wyndmoor, PA, United States of America
- * E-mail:
| | - Brian ByongKwon Yoo
- Centers for Disease Control and Prevention, Atlanta, GA, United States of America
| | - Cheng-An Hwang
- Eastern Regional Research Center, Agricultural Research Service, U.S. Department of Agriculture, Wyndmoor, PA, United States of America
| | - Mira Rakic Martinez
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States of America
| | - Atin R. Datta
- Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, MD, United States of America
| | - Pina M. Fratamico
- Eastern Regional Research Center, Agricultural Research Service, U.S. Department of Agriculture, Wyndmoor, PA, United States of America
| |
Collapse
|
24
|
Lian L, Li W, Xue T, Ren J, Tang F, Liu Y, Xue F, Dai J. Comparative transcriptomic analysis provides insights into transcription mechanisms of Vibrio parahaemolyticus T3SS during interaction with HeLa cells. Braz J Microbiol 2022; 53:289-301. [PMID: 34652743 PMCID: PMC8882520 DOI: 10.1007/s42770-021-00627-8] [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: 01/16/2021] [Accepted: 09/27/2021] [Indexed: 10/20/2022] Open
Abstract
Vibrio parahaemolyticus is an important foodborne pathogenic bacterium that harbors the type III secretion system 1 (T3SS1) as an essential virulence factor. However, the pathogenesis and infection mechanism mediated by T3SS1 are not entirely clarified. Similar to previous studies on other T3SS-positive bacteria, the T3SS1 needle is a major extracellular component in V. parahaemolyticus. We recently showed that the needle gene-deletion mutant (ΔvscF) exhibited markedly decreased cytotoxicity and effector translocation during interaction with HeLa cells. To further elucidate the pathogenesis of T3SS1 during host cell infection, bacterial RNA was extracted from wild-type POR-1 and ΔvscF mutants under infected condition for comparative RNA sequencing analysis in HeLa cell. The results showed that 120 differentially expressed genes (DEGs) were identified in the ΔvscF-infected group. These encoded proteins of DEGs, such as VP2088, VP2089, and VP2091, were annotated as ABC transporter system, whereas VP0757, VP1123, and VP1289 may be new transcriptional regulators. In addition, the downregulation of T3SS1 had a positive influence on the expression of T3SS2. Moreover, the transcription of the basal body is unaffected by the needle, and there was a close relation among the tip, translocon, and needle, because bacterial adenylate cyclase two-hybrid system (BACTH system) assay indicated the interaction of VP1656, VP1670, VP1693, and VP1694 (VscF). This study provides insights into transcription mechanism of T3SS1 upon infecting HeLa cell, which is expected to better clarify the T3SS1 virulent mechanism.
Collapse
Affiliation(s)
- Lele Lian
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Wanjun Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Tingyue Xue
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jianluan Ren
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Fang Tang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yongjie Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Feng Xue
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Jianjun Dai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
25
|
A Review on Transcriptional Responses of Interactions between Insect Vectors and Plant Viruses. Cells 2022; 11:cells11040693. [PMID: 35203347 PMCID: PMC8870222 DOI: 10.3390/cells11040693] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/03/2022] [Accepted: 02/15/2022] [Indexed: 02/04/2023] Open
Abstract
This review provides a synopsis of transcriptional responses pertaining to interactions between plant viruses and the insect vectors that transmit them in diverse modes. In the process, it attempts to catalog differential gene expression pertinent to virus–vector interactions in vectors such as virus reception, virus cell entry, virus tissue tropism, virus multiplication, and vector immune responses. Whiteflies, leafhoppers, planthoppers, and thrips are the main insect groups reviewed, along with aphids and leaf beetles. Much of the focus on gene expression pertinent to vector–virus interactions has centered around whole-body RNA extraction, whereas data on virus-induced tissue-specific gene expression in vectors is limited. This review compares transcriptional responses in different insect groups following the acquisition of non-persistent, semi-persistent, and persistent (non-propagative and propagative) plant viruses and identifies parallels and divergences in gene expression patterns. Understanding virus-induced changes in vectors at a transcriptional level can aid in the identification of candidate genes for targeting with RNAi and/or CRISPR editing in insect vectors for management approaches.
Collapse
|
26
|
Jovel J, Nimaga A, Jordan T, O’Keefe S, Patterson J, Thiesen A, Hotte N, Bording-Jorgensen M, Subedi S, Hamilton J, Carpenter EJ, Lauga B, Elahi S, Madsen KL, Wong GKS, Mason AL. Metagenomics Versus Metatranscriptomics of the Murine Gut Microbiome for Assessing Microbial Metabolism During Inflammation. Front Microbiol 2022; 13:829378. [PMID: 35185850 PMCID: PMC8851394 DOI: 10.3389/fmicb.2022.829378] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 01/11/2022] [Indexed: 01/26/2023] Open
Abstract
Shotgun metagenomics studies have improved our understanding of microbial population dynamics and have revealed significant contributions of microbes to gut homeostasis. They also allow in silico inference of the metagenome. While they link the microbiome with metabolic abnormalities associated with disease phenotypes, they do not capture microbial gene expression patterns that occur in response to the multitude of stimuli that constantly ambush the gut environment. Metatranscriptomics closes that gap, but its implementation is more expensive and tedious. We assessed the metabolic perturbations associated with gut inflammation using shotgun metagenomics and metatranscriptomics. Shotgun metagenomics detected changes in abundance of bacterial taxa known to be SCFA producers, which favors gut homeostasis. Bacteria in the phylum Firmicutes were found at decreased abundance, while those in phyla Bacteroidetes and Proteobacteria were found at increased abundance. Surprisingly, inferring the coding capacity of the microbiome from shotgun metagenomics data did not result in any statistically significant difference, suggesting functional redundancy in the microbiome or poor resolution of shotgun metagenomics data to profile bacterial pathways, especially when sequencing is not very deep. Obviously, the ability of metatranscriptomics libraries to detect transcripts expressed at basal (or simply low) levels is also dependent on sequencing depth. Nevertheless, metatranscriptomics informed about contrasting roles of bacteria during inflammation. Functions involved in nutrient transport, immune suppression and regulation of tissue damage were dramatically upregulated, perhaps contributed by homeostasis-promoting bacteria. Functions ostensibly increasing bacteria pathogenesis were also found upregulated, perhaps as a consequence of increased abundance of Proteobacteria. Bacterial protein synthesis appeared downregulated. In summary, shotgun metagenomics was useful to profile bacterial population composition and taxa relative abundance, but did not inform about differential gene content associated with inflammation. Metatranscriptomics was more robust for capturing bacterial metabolism in real time. Although both approaches are complementary, it is often not possible to apply them in parallel. We hope our data will help researchers to decide which approach is more appropriate for the study of different aspects of the microbiome.
Collapse
Affiliation(s)
- Juan Jovel
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
- Office of Research, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Juan Jovel,
| | - Aissata Nimaga
- Universite de Pau et des Pays de l’Adour, E2S UPPA, CNRS, IPREM, Pau, France
| | - Tracy Jordan
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Sandra O’Keefe
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Jordan Patterson
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Aducio Thiesen
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Naomi Hotte
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | | | - Sudip Subedi
- Office of Research, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Jessica Hamilton
- Office of Research, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Eric J. Carpenter
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Béatrice Lauga
- Universite de Pau et des Pays de l’Adour, E2S UPPA, CNRS, IPREM, Pau, France
| | - Shokrollah Elahi
- School of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Karen L. Madsen
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Gane Ka-Shu Wong
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
- BGI-Shenzhen, Beishan Industrial Zone, Shenzhen, China
| | - Andrew L. Mason
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
- Andrew L. Mason,
| |
Collapse
|
27
|
Serrano FA, Yukl ET. Contributions of Conformational Flexibility to High-Affinity Zinc Binding in the Solute Binding Protein AztC. ACS OMEGA 2022; 7:3768-3774. [PMID: 35128285 PMCID: PMC8811889 DOI: 10.1021/acsomega.1c06639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/10/2022] [Indexed: 06/14/2023]
Abstract
Bacteria rely on ATP binding cassette (ABC) transporters for the import of various nutrients. Bacterial ABC importers utilize an extracellular solute binding protein (SBP) to bind the substrate with high affinity and specificity and deliver it to the membrane permease for transport. The essential metals iron, manganese, and zinc are bound and transported by the cluster A-I SBPs. Crystal structures exist for the metal-bound and metal-free forms of several cluster A-I SBPs that show relatively subtle conformational changes that accompany metal binding. Recent solution studies and molecular dynamics simulations indicate a more complex conformational landscape for the cluster A-I SBPs, suggesting that changes in protein dynamics upon metal binding may have an important role in recognition by the membrane permease and effective transport. Here, we investigate conformational states and dynamics in the cluster A-I SBP AztC fromParacoccus denitrificans, characterizing its unusual intrinsic fluorescence behavior and thermodynamics of zinc binding. These data suggest a dynamic equilibrium of at least two conformational states in the apo form and compensatory changes in the holo that provide for a significant entropic contribution to zinc binding. Correlation with available crystal structures suggests that the formation of a Trp-Phe π-stacking interaction in the metal-bound form may mediate the observed changes in fluorescence. The conformational dynamics identified here for AztC are likely applicable to other cluster A-I SBPs with relevance to their exploitation as potential antibiotic drug targets.
Collapse
Affiliation(s)
- Fred A. Serrano
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico 88003, United States
| | - Erik T. Yukl
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, New Mexico 88003, United States
| |
Collapse
|
28
|
Niche differentiation of belowground microorganisms and their functional signatures in Assam type tea (Camellia sinensis var. assamica). Arch Microbiol 2021; 203:5661-5674. [PMID: 34462787 DOI: 10.1007/s00203-021-02547-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/10/2021] [Accepted: 08/19/2021] [Indexed: 12/29/2022]
Abstract
We employed an Illumina-based high-throughput metagenomics sequencing approach to unveil the rhizosphere and root endosphere microbial community associated with an organically grown Camellia population located at the Experimental Garden for Plantation Crops, Assam (India). The de novo assembled tea root endosphere metagenome contained 24,231 contigs (total 7,771,089 base pairs with an average length of 321 bps), while tea rhizosphere soil metagenome contained 261,965 sequences (total 230,537,174 base pairs, average length 846). The most prominent rhizobacteria belonged to the genera, viz., Bacillus (10.35%), Candidatus Solibacter (6.36%), Burkholderia (5.19%), Pseudomonas (3.9%), Streptomyces (3.52%), and Bradyrhizobium (2.77%), while the root endosphere was dominated by bacterial genera, viz., Serratia (46.64%), Methylobacterium (8.02%), Yersinia (5.97%), Burkholderia (2.05%), etc. The presence of few agronomically important bacterial genera, Bradyrhizobium, Rhizobium (each 0.93%), Sinorhizobium (0.34%), Azorhizobium, and Flavobacterium (0.17% each), was also detected in the root endosphere. KEGG pathway mapping indicated the presence of microbial metabolic pathway genes related to tyrosine metabolism, tryptophan metabolism, glyoxylate, and dicarboxylate metabolism which play important roles in endosphere activities, including survival, growth promotion, and host adaptation. The root endosphere microbiome also contained few important plant growth promoting traits related to phytohormone production, abiotic stress alleviation, mineral solubilization, and plant disease suppression.
Collapse
|
29
|
Thélot FA, Zhang W, Song K, Xu C, Huang J, Liao M. Distinct allosteric mechanisms of first-generation MsbA inhibitors. Science 2021; 374:580-585. [PMID: 34554829 DOI: 10.1126/science.abi9009] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- François A Thélot
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.,Biological and Biomedical Sciences Program, Harvard University, Cambridge, MA, USA
| | - Wenyi Zhang
- Key Laboratory of Structural Biology of Zhejiang Province, Westlake University, Hangzhou, China.,Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - KangKang Song
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA.,Cryo-EM Core Facility, University of Massachusetts Medical School, Worcester, MA, USA
| | - Chen Xu
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA.,Cryo-EM Core Facility, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jing Huang
- Key Laboratory of Structural Biology of Zhejiang Province, Westlake University, Hangzhou, China.,Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Maofu Liao
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Li Y, Ariotti N, Aghaei B, Pandzic E, Ganda S, Willcox M, Sanchez‐Felix M, Stenzel M. Inhibition of
S. aureus
Infection of Human Umbilical Vein Endothelial Cells (HUVECs) by Trehalose‐ and Glucose‐Functionalized Gold Nanoparticles. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202106544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Yimeng Li
- Centre for Advanced Macromolecular Design School of Chemistry University of New South Wales Sydney NSW 2052 Australia
| | - Nicholas Ariotti
- Electron Microscope Unit Mark Wainwright Analytical Centre University of New South Wales Sydney NSW 2052 Australia
| | - Behnaz Aghaei
- Inventia Life Science Pty Ltd Sydney NSW 2015 Australia
- School of Biotechnology and Biomolecular Sciences University of New South Wales Sydney NSW 2052 Australia
| | - Elvis Pandzic
- Katharina Gaus Light Microscopy Facility Mark Wainwright Analytical Centre University of New South Wales Sydney NSW 2052 Australia
| | - Sylvia Ganda
- Centre for Advanced Macromolecular Design School of Chemistry University of New South Wales Sydney NSW 2052 Australia
| | - Mark Willcox
- School of Optometry and Vision Science University of New South Wales Sydney NSW 2052 Australia
| | | | - Martina Stenzel
- Centre for Advanced Macromolecular Design School of Chemistry University of New South Wales Sydney NSW 2052 Australia
| |
Collapse
|
31
|
Župan ML, Luo Z, Ganio K, Pederick VG, Neville SL, Deplazes E, Kobe B, McDevitt CA. Conformation of the Solute-Binding Protein AdcAII Influences Zinc Uptake in Streptococcus pneumoniae. Front Cell Infect Microbiol 2021; 11:729981. [PMID: 34490149 PMCID: PMC8416893 DOI: 10.3389/fcimb.2021.729981] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 07/27/2021] [Indexed: 01/13/2023] Open
Abstract
Streptococcus pneumoniae scavenges essential zinc ions from the host during colonization and infection. This is achieved by the ATP-binding cassette transporter, AdcCB, and two solute-binding proteins (SBPs), AdcA and AdcAII. It has been established that AdcAII serves a greater role during initial infection, but the molecular details of how the protein selectively acquires Zn(II) remain poorly understood. This can be attributed to the refractory nature of metal-free AdcAII to high-resolution structural determination techniques. Here, we overcome this issue by separately mutating the Zn(II)-coordinating residues and performing a combination of structural and biochemical analyses on the variant proteins. Structural analyses of Zn(II)-bound AdcAII variants revealed that specific regions within the protein underwent conformational changes via direct coupling to each of the metal-binding residues. Quantitative in vitro metal-binding assays combined with affinity determination and phenotypic growth assays revealed that each of the four Zn(II)-coordinating residues contributes to metal binding by AdcAII. Intriguingly, the phenotypic growth impact of the mutant adcAII alleles was, in general, independent of affinity, suggesting that the Zn(II)-bound conformation of the SBP is crucial for efficacious metal uptake. Collectively, these data highlight the intimate coupling of ligand affinity with protein conformational change in ligand-receptor proteins and provide a putative mechanism for AdcAII. These findings provide further mechanistic insight into the structural and functional diversity of SBPs that is broadly applicable to other prokaryotes.
Collapse
Affiliation(s)
- Marina L Župan
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Zhenyao Luo
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Katherine Ganio
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Victoria G Pederick
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Stephanie L Neville
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Evelyne Deplazes
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, Australia.,School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Boštjan Kobe
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Christopher A McDevitt
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
32
|
Li Y, Ariotti N, Aghaei B, Pandzic E, Ganda S, Willcox M, Sanchez-Felix M, Stenzel MH. Inhibition of S. aureus-Infection of HUVECs by Trehalose and Glucose-functionalized Gold Nanoparticles. Angew Chem Int Ed Engl 2021; 60:22652-22658. [PMID: 34387412 DOI: 10.1002/anie.202106544] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/19/2021] [Indexed: 11/10/2022]
Abstract
Microbial adhesion to host cells represents the initial step in the infection process. Several methods have been explored to inhibit microbial adhesion including the use of glycopolymers based on mannose, galactose, sialic acid and glucose. These sugar receptors are however abundant in the body and they are not unique to bacteria. Trehalose in contrast is a unique disaccharide that is wildly expressed by microbes. This carbohydrate has not yet been explored as an anti-adhesive. Herein, gold nanoparticles (AuNPs) coated with trehalose-based polymers were prepared and compared to glucose-functionalized AuNPs and examined for their ability to prevent binding to endothelial cells. Acting as anti-adhesive, trehalose-functionalized nanoparticles decreased the binding of S. aureus to HUVEC cells, while outperforming the control nanoparticles. Microscopy revealed that trehalose coated nanoparticle bound strongly to S. aureus compared to the controls. In conclusion, nanoparticles based on trehalose could be a non-toxic alternative to inhibit S. aureus infection.
Collapse
Affiliation(s)
- Yimeng Li
- University of New South Wales - Kensington Campus: University of New South Wales, School of Chemistry, AUSTRALIA
| | - Nicholas Ariotti
- University of New South Wales - Kensington Campus: University of New South Wales, Mark Wainwright Analytical Centre, AUSTRALIA
| | - Behnaz Aghaei
- UNSW: University of New South Wales, school of Chemistry, AUSTRALIA
| | - Elvis Pandzic
- UNSW: University of New South Wales, school of chemistry, AUSTRALIA
| | - Sylvia Ganda
- UNSW: University of New South Wales, School of Chemistry, AUSTRALIA
| | - Mark Willcox
- UNSW: University of New South Wales, School of Optometry and Vision Science, AUSTRALIA
| | | | - Martina Heide Stenzel
- University of New South Wales Institute of Languages: UNSW Global Pty Limited, School of Chemical Sciences and Engineering, Applied Science Building, 2052, Sydney, AUSTRALIA
| |
Collapse
|
33
|
Correlation Analysis between Gut Microbiota and Metabolites in Children with Systemic Lupus Erythematosus. J Immunol Res 2021; 2021:5579608. [PMID: 34341764 PMCID: PMC8325587 DOI: 10.1155/2021/5579608] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/30/2021] [Accepted: 07/02/2021] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune-mediated diffuse connective tissue disease characterized by immune inflammation with an unclear aetiology and pathogenesis. This work profiled the intestinal flora and faecal metabolome of patients with SLE using 16S RNA sequencing and gas chromatography-mass spectrometry (GC-MS). We identified unchanged alpha diversity and partially altered beta diversity of the intestinal flora. Another important finding was the increase in Proteobacteria and Enterobacteriales and the decrease in Ruminococcaceae among SLE patients. For metabolites, amino acids and short-chain fatty acids were enriched when long-chain fatty acids were downregulated in SLE faecal samples. KEGG analysis showed the significance of the protein digestion and absorption pathway, and association analysis revealed the key role of 3-phenylpropanoic acid and Sphingomonas. Sphingomonas were reported to be less abundant in healthy periodontal sites of SLE patients than in those of HCs, indicating transmission of oral species to the gut. This study contributes to the understanding of the pathogenesis of SLE disease from the perspective of intestinal microorganisms, explains the pathogenesis of SLE, and serves as a basis for exploring potential treatments for the disease.
Collapse
|
34
|
Yang J, He C, Zhang H, Liu M, Zhao H, Ren L, Wu D, Du F, Liu B, Han X, He S, Chen Z. Evaluation and Differential Diagnosis of a Genetic Marked Brucella Vaccine A19ΔvirB12 for Cattle. Front Immunol 2021; 12:679560. [PMID: 34163479 PMCID: PMC8215367 DOI: 10.3389/fimmu.2021.679560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/10/2021] [Indexed: 01/23/2023] Open
Abstract
Brucella abortus is an important zoonotic pathogen that causes severe economic loss to husbandry and poses a threat to human health. The B. abortus A19 live vaccine has been extensively used to prevent bovine brucellosis in China. However, it is difficult to distinguish the serological response induced by A19 from that induced by natural infection. In this study, a novel genetically marked vaccine, A19ΔvirB12, was generated and evaluated. The results indicated that A19ΔvirB12 was able to provide effective protection against B. abortus 2308 (S2308) challenge in mice. Furthermore, the safety and protective efficacy of A19ΔvirB12 have been confirmed in natural host cattle. Additionally, the VirB12 protein allowed for serological differentiation between the S2308 challenge/natural infection and A19ΔvirB12 vaccination. However, previous studies have found that the accuracy of the serological detection based on VirB12 needs to be improved. Therefore, we attempted to identify potential supplementary antigens with differential diagnostic functions by combining label-free quantitative proteomics and protein chip technology. Twenty-six proteins identified only in S2308 were screened; among them, five proteins were considered as potential supplementary antigens. Thus, the accuracy of the differential diagnosis between A19ΔvirB12 immunization and field infection may be improved through multi-antigen detection. In addition, we explored the possible attenuation factors of Brucella vaccine strain. Nine virulence factors were downregulated in A19ΔvirB12. The downregulation pathways of A19ΔvirB12 were significantly enriched in quorum sensing, ATP-binding cassette transporter, and metabolism. Several proteins related to cell division were significantly downregulated, while some proteins involved in transcription were upregulated in S2308. In conclusion, our results contribute to the control and eradication of brucellosis and provide insights into the mechanisms underlying the attenuation of A19ΔvirB12.
Collapse
Affiliation(s)
- Jianghua Yang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Chuanyu He
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China.,Tecon Biological Co. Ltd., Urumqi, China
| | - Huan Zhang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | | | | | - Lisong Ren
- Tecon Biological Co. Ltd., Urumqi, China
| | | | - Fangyuan Du
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Baoshan Liu
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Xiaohu Han
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China
| | - Sun He
- Tecon Biological Co. Ltd., Urumqi, China
| | - Zeliang Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China.,Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China.,Brucellosis Prevention and Treatment Engineering Technology Research Center of Inner Mongolia Autonomous Region, Inner Mongolia University for Nationalities, Tongliao, China.,School of Public Health, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
35
|
Potential factors involved in the early pathogenesis of Streptococcus uberis mastitis: a review. Folia Microbiol (Praha) 2021; 66:509-523. [PMID: 34085166 DOI: 10.1007/s12223-021-00879-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 05/24/2021] [Indexed: 10/21/2022]
Abstract
Bovine mastitis is an inflammation of the mammary gland, which could be the result of allergy, physical trauma, or invasion by pathogens as Streptococcus uberis. This pathogen is an environmental pathogen associated with subclinical and clinical intramammary infection (IMI) in both lactating and non-lactating cows, which can persist in the udder and cause a chronic infection in the mammary gland. In spite of the important economic losses and increased prevalence caused by S. uberis mastitis, virulence factors involved in bacterial colonization of mammary glands and the pathogenic mechanisms are not yet clear. In the last 30 years, several studies have defined adherence and internalization of S. uberis as the early stages in IMI. S. uberis adheres to and invades into mammary gland cells, and this ability has been observed in in vitro assays. Until now, these abilities have not been determined in vivo challenges since they have been difficult to study. Bacterial surface proteins are able to bind to extracellular matrix protein components such as fibronectin, collagen and laminin, as well as proteins in milk. These proteins play a role in adhesion to host cells and have been denominated microbial surface components recognizing adhesive matrix molecules (MSCRAMMs). This article aims to summarize our current knowledge on the most relevant properties of the potential factors involved in the early pathogenesis of S. uberis mastitis.
Collapse
|
36
|
Pandey A, Boros E. Coordination Complexes to Combat Bacterial Infections: Recent Developments, Current Directions and Future Opportunities. Chemistry 2021; 27:7340-7350. [PMID: 33368662 DOI: 10.1002/chem.202004822] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/22/2020] [Indexed: 12/29/2022]
Abstract
Drug discovery aimed at the efficient eradication of life-threatening bacterial infections, especially in light of the emergence of multi-drug resistance of pathogenic bacteria, has remained a challenge for medicinal chemists over the past several decades. As nutrient acquisition and metabolism at the host-pathogen interface become better elucidated, new drug targets continue to emerge. Metal homeostasis is among these processes, and thus provides opportunities for medicinal inorganic chemists to alter or disrupt these processes selectively to impart bacteriostatic or bacteriotoxic effects. In this minireview, we showcase some of the recent work from the field of metal-based antibacterial agents and highlight divergent strategies and mechanisms of action.
Collapse
Affiliation(s)
- Apurva Pandey
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, NY, 11794, USA
| | - Eszter Boros
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, NY, 11794, USA
| |
Collapse
|
37
|
Qiu X, Zhang Y, Hong H. Classification of acetic acid bacteria and their acid resistant mechanism. AMB Express 2021; 11:29. [PMID: 33595734 PMCID: PMC7889782 DOI: 10.1186/s13568-021-01189-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 02/04/2021] [Indexed: 12/14/2022] Open
Abstract
Acetic acid bacteria (AAB) are obligate aerobic Gram-negative bacteria that are commonly used in vinegar fermentation because of their strong capacity for ethanol oxidation and acetic acid synthesis as well as their acid resistance. However, low biomass and low production rate due to acid stress are still major challenges that must be overcome in industrial processes. Although acid resistance in AAB is important to the production of high acidity vinegar, the acid resistance mechanisms of AAB have yet to be fully elucidated. In this study, we discuss the classification of AAB species and their metabolic processes and review potential acid resistance factors and acid resistance mechanisms in various strains. In addition, we analyze the quorum sensing systems of Komagataeibacter and Gluconacetobacter to provide new ideas for investigation of acid resistance mechanisms in AAB in the form of signaling pathways. The results presented herein will serve as an important reference for selective breeding of high acid resistance AAB and optimization of acetic acid fermentation processes.
Collapse
Affiliation(s)
- Xiaoman Qiu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, No. 30, Puzhu Road, Nanjing, 211800, China
- National Engineering Technique Research Center for Biotechnology, Nanjing Tech University, No. 30, Puzhu Road, Nanjing, 211800, China
| | - Yao Zhang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, No. 30, Puzhu Road, Nanjing, 211800, China
- National Engineering Technique Research Center for Biotechnology, Nanjing Tech University, No. 30, Puzhu Road, Nanjing, 211800, China
| | - Housheng Hong
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, No. 30, Puzhu Road, Nanjing, 211800, China.
- National Engineering Technique Research Center for Biotechnology, Nanjing Tech University, No. 30, Puzhu Road, Nanjing, 211800, China.
| |
Collapse
|
38
|
Abstract
Zinc is an essential nutrient for the virulence of bacterial pathogens such as Streptococcus pneumoniae. Many Gram-positive bacteria use a two-domain lipoprotein for zinc acquisition, but how this class of metal-recruiting proteins acquire zinc and interact with the uptake machinery has remained poorly defined. Zinc is an essential element in all domains of life. Nonetheless, how prokaryotes achieve selective acquisition of zinc from the extracellular environment remains poorly understood. Here, we elucidate a novel mechanism for zinc-binding in AdcA, a solute-binding protein of Streptococcus pneumoniae. Crystal structure analyses reveal the two-domain organization of the protein and show that only the N-terminal domain (AdcAN) is necessary for zinc import. Zinc binding induces only minor changes in the global protein conformation of AdcA and stabilizes a highly mobile loop within the AdcAN domain. This loop region, which is conserved in zinc-specific solute-binding proteins, facilitates closure of the AdcAN binding site and is crucial for zinc acquisition. Collectively, these findings elucidate the structural and functional basis of selective zinc uptake in prokaryotes.
Collapse
|
39
|
Yan R, Jiang H, Gu S, Feng N, Zhang N, Lv L, Liu F. Fecal Metabolites Were Altered, Identified as Biomarkers and Correlated With Disease Activity in Patients With Systemic Lupus Erythematosus in a GC-MS-Based Metabolomics Study. Front Immunol 2020; 11:2138. [PMID: 33013903 PMCID: PMC7511511 DOI: 10.3389/fimmu.2020.02138] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/06/2020] [Indexed: 12/11/2022] Open
Abstract
Gut metabolites are products of the crosstalk between microbes and their host and play an important role in the occurrence, development, diagnosis, and treatment of autoimmune diseases. This work profiled the fecal metabolome of patients with systemic lupus erythematosus (SLE) using gas chromatography-mass spectrometry (GC-MS) and analyzed the potential roles of metabolites in the diagnosis and development of SLE. Fecal sample from 29 SLE patients without any other diseases and 30 healthy controls (HCs) were analyzed by metabolomics profiling. All participants took no antibiotics in the month before sampling and clinical data collecting. The metabolome profiles of patients with SLE and HCs were significantly different. Thirty fecal metabolites, such as deoxycholic acid, erucamide, L-tryptophan and putrescine, were significantly enriched, while nine metabolites, such as glyceric acid, γ-tocopherol, (Z)-13-octadecenoic acid and 2,4-di-tert-butylphenol, were depleted in SLE patients vs. HCs. The areas under the curve (AUCs) of L-valine, pyrimidine, erucamide, and L-leucine during ROC analysis were 0.886, 0.833, 0.829, and 0.803, indicating their good diagnostic potential. Moreover, the combination of L-valine, erucamide and 2,4-di-tert-butylphenol gave an AUC of 0.959. SLE-altered metabolites were significantly located in 28 pathways, such as ABC transporters (p = 3.40E-13) and aminoacyl-tRNA biosynthesis (p = 2.11E-12). Furthermore, SLE-altered fecal metabolites were closely correlated with SLE indicators, e.g., L-tryptophan was positively correlated with the SLEDAI-2K (p = 0.007). Our results suggest that the SLE fecal metabolome is closely associated with the occurrence and development of SLE and is of great diagnostic value.
Collapse
Affiliation(s)
- Ren Yan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Huiyong Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Silan Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ninghan Feng
- Department of Urology, Affiliated Wuxi No.2 People's Hospital, Nanjing Medical University, Wuxi, China
| | - Nan Zhang
- Department of Urology, Affiliated Wuxi No.2 People's Hospital, Nanjing Medical University, Wuxi, China
| | - Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Fengping Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
40
|
Grechko V, Podolsky D, Cheshchevik V. Identification new potential multidrug resistance proteins of Saccharomyces cerevisiae. J Microbiol Methods 2020; 176:106029. [DOI: 10.1016/j.mimet.2020.106029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/28/2020] [Accepted: 08/06/2020] [Indexed: 10/23/2022]
|
41
|
Xie ZY, Gong XX, Xu XD, Mei B, Xuan XZ, Long H, Zhang X, Cai XN. Identification of Vibrio alginolyticus virulent strain-specific DNA regions by suppression subtractive hybridization and PCR. J Appl Microbiol 2020; 129:1472-1485. [PMID: 32510751 DOI: 10.1111/jam.14739] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/23/2020] [Accepted: 06/02/2020] [Indexed: 12/25/2022]
Abstract
AIMS Vibrio alginolyticus was frequently isolated from diseased farmed fish in the coaster waters of Hainan Island over the past two decades. In this study, we attempted to identify candidates of virulent strain-specific DNA regions for this pathogen. METHODS AND RESULTS Suppression subtractive hybridization (SSH) and PCR were successively performed between the typical virulent strain and avirulent strain of V. alginolyticus, in which they shared 99·54% homology of 16S rDNAs. Out of 2873 subtracted clones, nine clones were finally indicated to harbour virulent strain-specific DNA fragments. The receivable functions of the major fragments in the nine clones were believed to encode methyl-accepting chemotaxis protein (n = 1), type VI secretion system-associated FHA domain protein TagH (n = 1), diguanylate cyclase (n = 1), AraC family transcriptional regulator (n = 1), ABC-type uncharacterized transport system permease component (n = 1) and hypothetical proteins (n = 4). Two hypothetical proteins contain several disordered regions. CONCLUSIONS Some specific DNA regions existed in the virulent strain of V. alginolyticus, and the SSH assay could be a highly sensitive method for identifying virulent regions in pathogens. SIGNIFICANCE AND IMPACT OF THE STUDY This report is the first to describe the identification of virulent strain-specific DNA regions in the V. alginolyticus genome, which is helpful in developing virulent strain-specific rapid detection methods and is a pivotal precondition for clarifying the molecular virulence mechanism of V. alginolyticus.
Collapse
Affiliation(s)
- Z-Y Xie
- State Key Laboratory of Marine Resource Utilization in the South China Sea, Hainan University, Haikou, Hainan Province, China.,Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, Hainan University, Haikou, Hainan Province, China.,College of Marine Sciences, Hainan University, Haikou, Hainan Province, China
| | - X-X Gong
- State Key Laboratory of Marine Resource Utilization in the South China Sea, Hainan University, Haikou, Hainan Province, China.,Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, Hainan University, Haikou, Hainan Province, China.,College of Marine Sciences, Hainan University, Haikou, Hainan Province, China
| | - X-D Xu
- College of Marine Sciences, Hainan University, Haikou, Hainan Province, China
| | - B Mei
- College of Marine Sciences, Hainan University, Haikou, Hainan Province, China
| | - X-Z Xuan
- College of Marine Sciences, Hainan University, Haikou, Hainan Province, China
| | - H Long
- State Key Laboratory of Marine Resource Utilization in the South China Sea, Hainan University, Haikou, Hainan Province, China
| | - X Zhang
- State Key Laboratory of Marine Resource Utilization in the South China Sea, Hainan University, Haikou, Hainan Province, China.,Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, Hainan University, Haikou, Hainan Province, China.,College of Marine Sciences, Hainan University, Haikou, Hainan Province, China
| | - X-N Cai
- State Key Laboratory of Marine Resource Utilization in the South China Sea, Hainan University, Haikou, Hainan Province, China.,Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, Hainan University, Haikou, Hainan Province, China.,College of Marine Sciences, Hainan University, Haikou, Hainan Province, China
| |
Collapse
|
42
|
Sabrialabed S, Yang JG, Yariv E, Ben-Tal N, Lewinson O. Substrate recognition and ATPase activity of the E. coli cysteine/cystine ABC transporter YecSC-FliY. J Biol Chem 2020; 295:5245-5256. [PMID: 32144203 PMCID: PMC7170509 DOI: 10.1074/jbc.ra119.012063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/03/2020] [Indexed: 12/20/2022] Open
Abstract
Sulfur is essential for biological processes such as amino acid biogenesis, iron-sulfur cluster formation, and redox homeostasis. To acquire sulfur-containing compounds from the environment, bacteria have evolved high-affinity uptake systems, predominant among which is the ABC transporter family. Theses membrane-embedded enzymes use the energy of ATP hydrolysis for transmembrane transport of a wide range of biomolecules against concentration gradients. Three distinct bacterial ABC import systems of sulfur-containing compounds have been identified, but the molecular details of their transport mechanism remain poorly characterized. Here we provide results from a biochemical analysis of the purified Escherichia coli YecSC-FliY cysteine/cystine import system. We found that the substrate-binding protein FliY binds l-cystine, l-cysteine, and d-cysteine with micromolar affinities. However, binding of the l- and d-enantiomers induced different conformational changes of FliY, where the l- enantiomer-substrate-binding protein complex interacted more efficiently with the YecSC transporter. YecSC had low basal ATPase activity that was moderately stimulated by apo FliY, more strongly by d-cysteine-bound FliY, and maximally by l-cysteine- or l-cystine-bound FliY. However, at high FliY concentrations, YecSC reached maximal ATPase rates independent of the presence or nature of the substrate. These results suggest that FliY exists in a conformational equilibrium between an open, unliganded form that does not bind to the YecSC transporter and closed, unliganded and closed, liganded forms that bind this transporter with variable affinities but equally stimulate its ATPase activity. These findings differ from previous observations for similar ABC transporters, highlighting the extent of mechanistic diversity in this large protein family.
Collapse
Affiliation(s)
- Siwar Sabrialabed
- Department of Biochemistry and the Rappaport Institute for Medical Sciences, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Janet G Yang
- Department of Chemistry, University of San Francisco, San Francisco, California 94117
| | - Elon Yariv
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6139001, Israel
| | - Nir Ben-Tal
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6139001, Israel
| | - Oded Lewinson
- Department of Biochemistry and the Rappaport Institute for Medical Sciences, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
43
|
Rathje K, Mortzfeld B, Hoeppner MP, Taubenheim J, Bosch TCG, Klimovich A. Dynamic interactions within the host-associated microbiota cause tumor formation in the basal metazoan Hydra. PLoS Pathog 2020; 16:e1008375. [PMID: 32191776 PMCID: PMC7081986 DOI: 10.1371/journal.ppat.1008375] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 02/01/2020] [Indexed: 02/07/2023] Open
Abstract
The extent to which disturbances in the resident microbiota can compromise an animal’s health is poorly understood. Hydra is one of the evolutionary oldest animals with naturally occurring tumors. Here, we found a causal relationship between an environmental spirochete (Turneriella spec.) and tumorigenesis in Hydra. Unexpectedly, virulence of this pathogen requires the presence of Pseudomonas spec., a member of Hydra´s beneficial microbiome indicating that dynamic interactions between a resident bacterium and a pathogen cause tumor formation. The observation points to the crucial role of commensal bacteria in maintaining tissue homeostasis and adds support to the view that microbial community interactions are essential for disease. These findings in an organism that shares deep evolutionary connections with all animals have implications for our understanding of cancer. Here we follow up on our initial observation of tumor formation in the basal metazoan Hydra and demonstrate that tumor development in one of the evolutionary oldest animals is caused by a dynamic interplay between an environmental spirochete, the host-associated resident microbiota, and the tissue homeostasis within the animal. Unexpectedly, the pathogenicity of the environmental bacterium Turneriella is context-dependent: the virulence of this pathogen requires the presence of a member of Hydra’s beneficial microbiome—the Pseudomonas bacterium. Dynamic interactions between two microbiota members have profound effects onto the host tissue homeostasis and fitness. Our data provide direct evidence for the important role of the resident microbiome in maintaining tissue homeostasis and pathogen defense, a fundamental process that is likely to take place in every tissue of every animal species. In summary, our study uncovers an evolutionary conserved role of the resident microbiome in guarding host’s tissue homeostasis.
Collapse
Affiliation(s)
- Kai Rathje
- Zoological Institute, Kiel University, Kiel, Germany
| | - Benedikt Mortzfeld
- Zoological Institute, Kiel University, Kiel, Germany
- Department of Biology, University of Massachusetts Dartmouth, Dartmouth, Massachusetts, United States of America
| | - Marc P. Hoeppner
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Jan Taubenheim
- Zoological Institute, Kiel University, Kiel, Germany
- Institute for Zoology and Organismic Interactions, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Thomas C. G. Bosch
- Zoological Institute, Kiel University, Kiel, Germany
- * E-mail: (TCGB); (AK)
| | | |
Collapse
|
44
|
Abstract
Energy-coupling factor (ECF)-type ATP-binding cassette (ABC) transporters catalyze membrane transport of micronutrients in prokaryotes. Crystal structures and biochemical characterization have revealed that ECF transporters are mechanistically distinct from other ABC transport systems. Notably, ECF transporters make use of small integral membrane subunits (S-components) that are predicted to topple over in the membrane when carrying the bound substrate from the extracellular side of the bilayer to the cytosol. Here, we review the phylogenetic diversity of ECF transporters as well as recent structural and biochemical advancements that have led to the postulation of conceptually different mechanistic models. These models can be described as power stroke and thermal ratchet. Structural data indicate that the lipid composition and bilayer structure are likely to have great impact on the transport function. We argue that study of ECF transporters could lead to generic insight into membrane protein structure, dynamics, and interaction.
Collapse
Affiliation(s)
- S Rempel
- Gr oningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, The Netherlands; , ,
| | - W K Stanek
- Gr oningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, The Netherlands; , ,
| | - D J Slotboom
- Gr oningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, 9747 AG Groningen, The Netherlands; , , .,Zernike Institute for Advanced Materials, University of Groningen, 9747 AG Groningen, The Netherlands
| |
Collapse
|
45
|
Shishpal P, Kasarpalkar N, Singh D, Bhor VM. Characterization of Gardnerella vaginalis membrane vesicles reveals a role in inducing cytotoxicity in vaginal epithelial cells. Anaerobe 2019; 61:102090. [PMID: 31442559 DOI: 10.1016/j.anaerobe.2019.102090] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 08/18/2019] [Accepted: 08/19/2019] [Indexed: 12/14/2022]
Abstract
Bacterial vaginosis (BV) is a common polymicrobial infection affecting women in the reproductive age and is associated with adverse obstetric and gynaecological outcomes. Gardnerella vaginalis is the most virulent anaerobic bacterial species predominantly associated with BV. However, a clear understanding of the mechanisms by which it contributes to the pathogenesis and persistence of BV is lacking. In this report, we demonstrate for the first time, the isolation of membrane vesicles (MVs) from G. vaginalis ATCC 14019. These MVs are approximately 120-260 nm in diameter. Proteomic characterization of the MVs by LC-MS/MS led to the identification of 417 proteins, including proteins involved in cellular metabolism as well as molecular chaperones and certain virulence factors. Immunoblot analysis of the MVs confirmed the presence of vaginolysin, the most well-characterized virulence factor of G. vaginalis. The exposure of the vaginal epithelial cells, VK2/E6E7 to the G. vaginalis MVs resulted in the internalization of the MVs. The MVs induced cytotoxicity and an increase in the levels of the pro-inflammatory cytokine, IL-8 in VK2 cells as well lysis of erythrocytes. The results of the study indicate that G. vaginalis MVs may be involved in the delivery of cytotoxic proteins and other virulence factors to the host cells and could thereby contribute towards enhancing the cellular damage associated with pathogenesis of BV.
Collapse
Affiliation(s)
- Parul Shishpal
- Department of Molecular Immunology and Microbiology, Indian Council of Medical Research-National Institute for Research in Reproductive Health (ICMR-NIRRH), J. M. Street, Parel, Mumbai, India
| | - Nandini Kasarpalkar
- Department of Molecular Immunology and Microbiology, Indian Council of Medical Research-National Institute for Research in Reproductive Health (ICMR-NIRRH), J. M. Street, Parel, Mumbai, India
| | - Dipty Singh
- Department of Neuroendocrinology and Transmission Electron Microscopy, Indian Council of Medical Research-National Institute for Research in Reproductive Health (ICMR-NIRRH), J. M. Street, Parel, Mumbai, India
| | - Vikrant M Bhor
- Department of Molecular Immunology and Microbiology, Indian Council of Medical Research-National Institute for Research in Reproductive Health (ICMR-NIRRH), J. M. Street, Parel, Mumbai, India.
| |
Collapse
|
46
|
Gao L, Ma Y, Li X, Zhang L, Zhang C, Chen Q, Zhao C. Research on the roles of genes coding ATP‐binding cassette transporters in
Porphyromonas gingivalis
pathogenicity. J Cell Biochem 2019; 121:93-102. [PMID: 31081181 DOI: 10.1002/jcb.28887] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Li Gao
- Department of Periodontology, Guanghua School of Stomatology, Hospital of Stomatology Sun Yat‐Sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Stomatology Guangzhou China
| | - Yuanyuan Ma
- Guangdong Provincial Key Laboratory of Stomatology Guangzhou China
- Department of Prosthodontics, Guanghua School of Stomatology, Hospital of Stomatology Sun Yat‐Sen University Guangzhou China
| | - Xiting Li
- Department of Periodontology, Guanghua School of Stomatology, Hospital of Stomatology Sun Yat‐Sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Stomatology Guangzhou China
| | - Liping Zhang
- Guangdong Provincial Key Laboratory of Stomatology Guangzhou China
| | - Chi Zhang
- Guangdong Provincial Key Laboratory of Stomatology Guangzhou China
| | - Qianying Chen
- Guangdong Provincial Key Laboratory of Stomatology Guangzhou China
| | - Chuanjiang Zhao
- Department of Periodontology, Guanghua School of Stomatology, Hospital of Stomatology Sun Yat‐Sen University Guangzhou China
- Guangdong Provincial Key Laboratory of Stomatology Guangzhou China
| |
Collapse
|
47
|
Grim KP, San Francisco B, Radin JN, Brazel EB, Kelliher JL, Párraga Solórzano PK, Kim PC, McDevitt CA, Kehl-Fie TE. The Metallophore Staphylopine Enables Staphylococcus aureus To Compete with the Host for Zinc and Overcome Nutritional Immunity. mBio 2017; 8:e01281-17. [PMID: 29089427 PMCID: PMC5666155 DOI: 10.1128/mbio.01281-17] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 09/27/2017] [Indexed: 12/18/2022] Open
Abstract
During infection, the host sequesters essential nutrients, such as zinc, to combat invading microbes. Despite the ability of the immune effector protein calprotectin to bind zinc with subpicomolar affinity, Staphylococcus aureus is able to successfully compete with the host for zinc. However, the zinc importers expressed by S. aureus remain unknown. Our investigations have revealed that S. aureus possesses two importers, AdcABC and CntABCDF, which are induced in response to zinc limitation. While AdcABC is similar to known zinc importers in other bacteria, CntABCDF has not previously been associated with zinc acquisition. Concurrent loss of the two systems severely impairs the ability of S. aureus to obtain zinc and grow in zinc-limited environments. Further investigations revealed that the Cnt system is responsible for the ability of S. aureus to compete with calprotectin for zinc in culture and contributes to acquisition of zinc during infection. The cnt locus also enables S. aureus to produce the broad-spectrum metallophore staphylopine. Similarly to the Cnt transporter, loss of staphylopine severely impairs the ability of S. aureus to resist host-imposed zinc starvation, both in culture and during infection. Further investigations revealed that together staphylopine and the Cnt importer function analogously to siderophore-based iron acquisition systems in order to facilitate zinc acquisition by S. aureus Analogous systems are found in a broad range of Gram-positive and Gram-negative bacterial pathogens, suggesting that this new type of zinc importer broadly contributes to the ability of bacteria to cause infection.IMPORTANCE A critical host defense against infection is the restriction of zinc availability. Despite the subpicomolar affinity of the immune effector calprotectin for zinc, Staphylococcus aureus can successfully compete for this essential metal. Here, we describe two zinc importers, AdcABC and CntABCDF, possessed by S. aureus, the latter of which has not previously been associated with zinc acquisition. The ability of S. aureus to compete with the host for zinc is dependent on CntABCDF and the metallophore staphylopine, both in culture and during infection. These results expand the mechanisms utilized by bacteria to obtain zinc, beyond Adc-like systems, and demonstrate that pathogens utilize strategies similar to siderophore-based iron acquisition to obtain other essential metals during infection. The staphylopine synthesis machinery is present in a diverse collection of bacteria, suggesting that this new family of zinc importers broadly contributes to the ability of numerous pathogens to cause infection.
Collapse
Affiliation(s)
- Kyle P Grim
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Brian San Francisco
- Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Jana N Radin
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Erin B Brazel
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Jessica L Kelliher
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Paola K Párraga Solórzano
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Departamento de Ciencias de la Vida, Universidad de las Fuerzas Armadas ESPE, Sangolquí, Ecuador
| | - Philip C Kim
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Christopher A McDevitt
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Thomas E Kehl-Fie
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
48
|
Perrone F, De Siena B, Muscariello L, Kendall SL, Waddell SJ, Sacco M. A Novel TetR-Like Transcriptional Regulator Is Induced in Acid-Nitrosative Stress and Controls Expression of an Efflux Pump in Mycobacteria. Front Microbiol 2017; 8:2039. [PMID: 29109706 PMCID: PMC5660060 DOI: 10.3389/fmicb.2017.02039] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/05/2017] [Indexed: 11/30/2022] Open
Abstract
Mycobacterium tuberculosis has the ability to survive inside macrophages under acid-nitrosative stress. M. tuberculosis Rv1685c and its ortholog in M. smegmatis, MSMEG_3765, are induced on exposure to acid-nitrosative stress. Both genes are annotated as TetR transcriptional regulators, a family of proteins that regulate a wide range of cellular activities, including multidrug resistance, carbon catabolism and virulence. Here, we demonstrate that MSMEG_3765 is co-transcribed with the upstream genes MSMEG_3762 and MSMEG_3763, encoding efflux pump components. RTq-PCR and GFP-reporter assays showed that the MSMEG_3762/63/65 gene cluster, and the orthologous region in M. tuberculosis (Rv1687c/86c/85c), was up-regulated in a MSMEG_3765 null mutant, suggesting that MSMEG_3765 acts as a repressor, typical of this family of regulators. We further defined the MSMEG_3765 regulon using genome-wide transcriptional profiling and used reporter assays to confirm that the MSMEG_3762/63/65 promoter was induced under acid-nitrosative stress. A putative 36 bp regulatory motif was identified upstream of the gene clusters in both M. smegmatis and M. tuberculosis and purified recombinant MSMEG_3765 protein was found to bind to DNA fragments containing this motif from both M. smegmatis and M. tuberculosis upstream regulatory regions. These results suggest that the TetR repressor MSMEG_3765/Rv1685c controls expression of an efflux pump with an, as yet, undefined role in the mycobacterial response to acid-nitrosative stress.
Collapse
Affiliation(s)
- Filomena Perrone
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche e Farmaceutiche, Università della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Barbara De Siena
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche e Farmaceutiche, Università della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Lidia Muscariello
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche e Farmaceutiche, Università della Campania "Luigi Vanvitelli", Caserta, Italy
| | - Sharon L Kendall
- Department of Pathobiology and Population Science, Royal Veterinary College, London, United Kingdom
| | - Simon J Waddell
- Wellcome Trust Brighton and Sussex Centre for Global Health Research, Brighton and Sussex Medical School, University of Sussex, Brighton, United Kingdom
| | - Margherita Sacco
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche e Farmaceutiche, Università della Campania "Luigi Vanvitelli", Caserta, Italy
| |
Collapse
|
49
|
Sampaio A, Pegos VR, Oshiro EE, Balan A. The periplasmic binding protein NrtT affects xantham gum production and pathogenesis in Xanthomonas citri. FEBS Open Bio 2017; 7:1499-1514. [PMID: 28979839 PMCID: PMC5623697 DOI: 10.1002/2211-5463.12281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/19/2017] [Accepted: 07/28/2017] [Indexed: 12/05/2022] Open
Abstract
In Xanthomonas citri, the bacterium that causes citrus canker, three ATP-binding cassette (ABC) transporters are known to be dedicated to the uptake of sulfur compounds. In this work, using functional, biophysical and structural methods, we showed that NrtT, a periplasmic component of the ABC transporter NrtCB, is an alkanesulfonate-binding protein and that the deletion of the nrtT gene affected xantham gum synthesis, adhesion and biofilm production, similarly to the phenotype obtained in the X. citri ssuA-knockout strain, in which the alkanesulfonate-binding protein SsuA is absent. Although NrtA and SsuA share similar ligands, the function of these proteins is not complementary. These results emphasize that organic-sulfur sources are directly involved with bacterial infection in vivo and are needed for pathogenesis in X. citri.
Collapse
Affiliation(s)
- Aline Sampaio
- Programa Interunidades em BiotecnologiaInstituto de Ciências BiomédicasUniversidade de São PauloUSPBrazil
- Laboratório Nacional de Biociências (LNBio)Centro de Pesquisas em Energia e Materiais (CNPEM)São PauloBrazil
| | - Vanessa Rodrigues Pegos
- Laboratório Nacional de Biociências (LNBio)Centro de Pesquisas em Energia e Materiais (CNPEM)São PauloBrazil
- Post‐Graduate Program in Genetics and Molecular BiologyInstitute of BiologyUniversity of Campinas UNICAMPCampinasSão PauloBrazil
| | - Elisa Emiko Oshiro
- Departmento de MicrobiologiaInstituto de Ciências BiomédicasUniversidade de São PauloBrazil
| | - Andrea Balan
- Departmento de MicrobiologiaInstituto de Ciências BiomédicasUniversidade de São PauloBrazil
| |
Collapse
|
50
|
Quantum chemical calculations of the active site of the solute-binding protein PsaA from Streptococcus pneumoniae explain electronic selectivity of metal binding. Struct Chem 2017. [DOI: 10.1007/s11224-017-1036-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|