1
|
Ma Q, Liu Y, Cen J, Wang Q, Chen M, Chen S, Zhang Z, Han K, Feng Z, Wu C, Shen J, Jiang H. Disrupting cross-adaptation in high-risk MRSA: Sanguinarine as a multi-effective stress sensitizer for environmental and food safety. JOURNAL OF HAZARDOUS MATERIALS 2025; 494:138586. [PMID: 40367773 DOI: 10.1016/j.jhazmat.2025.138586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 04/30/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) represents a significant public health concern owing to its formidable antibiotic resistance and robust capacity for biofilm formation. The cross-adaptation mechanism enables MRSA to develop tolerance to environmental stressors such as antibiotics, acid, heat and osmotic pressure, leading to the persistence infections and environmental contamination. The cross-adaptation mechanism enables MRSA to develop tolerance to environmental stressors, such as antibiotics, acid, heat and osmotic pressure, leading to the persistence infections and environmental contamination. Here, we identified 261 strains of S. aureus and 9 high-risk MRSA from the environment of dairy farms and raw milk. The natural product Sanguinarine (SAN), derived from feed additives, exhibits effective anti-MRSA and anti-biofilm activity. Notably, SAN enhances the sensitivity of MRSA to antibiotics, acid, heat, and osmotic pressure by disrupting the cross-adaptation mechanism. Mechanistic investigations revealed that SAN significantly reduces the transcriptional level of type I (dnaK, groEL, etc.) and type III (clpB, clpP, etc.) heat stress response genes while markedly upregulating type II (σB) gene. Furthermore, SAN upregulates Na+/H+ antiporters activity, F0F1-ATPase activity and purine metabolism, while broadly downregulating DNA damage repair genes and disrupting ribosomal function. Additionally, SAN induces non-synonymous mutations in key stress response factors ClpB/L, leading to a loss of conformational homeostasis. SAN elicits a distinct stress response compared to environmental stressors, weakening MRSA's resilience and demonstrating promising capabilities for MRSA clearance and biofilm inhibition. Overall, SAN provides an effective strategy for the clearance of high-risk MRSA and the assurance of public health security.
Collapse
Affiliation(s)
- Qiang Ma
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yuan Liu
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jianuo Cen
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Qiqi Wang
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Meinuo Chen
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shiqi Chen
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhimeng Zhang
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ke Han
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhiyue Feng
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Congming Wu
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Haiyang Jiang
- National Key Laboratory of Veterinary Public Health and Security, College of Veterinary Medicine, China Agricultural University, Beijing, China.
| |
Collapse
|
2
|
Wu Z, Lei X, Zhang Y, Wu S, Hou Z, Ma K, Pei H, Shang F, Xue T. The membrane protein DtpT plays an important role in biofilm formation and stress resistance in foodborne Staphylococcus aureus RMSA49. Food Res Int 2025; 208:116249. [PMID: 40263806 DOI: 10.1016/j.foodres.2025.116249] [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: 12/17/2024] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 04/24/2025]
Abstract
Staphylococcus aureus has been a major contributor to the contamination of dairy products and preserved foods due to its capacity for biofilm formation and strong resistance to environmental stress. The membrane transport protein di-and tripeptides transporter (DtpT) is the primary transporter of di- and tripeptides in S. aureus, yet its impact on biofilm formation and stress resistance in S. aureus has not been previously reported. Our study focused on the foodborne S. aureus strain RMSA49, revealing that mutation of the dtpT resulted in diminished biofilm formation ability and reduced tolerance to environmental stress (high temperature, dryness, oxidative stress, and salt stress). These findings highlight the significance of DtpT in both biofilm formation and response to environmental stress in foodborne S. aureus. Our study represents the first report demonstrating the crucial role of DtpT in biofilm formation and environmental tolerance in S. aureus, providing new avenues for future research on this protein while also identifying potential target genes for further investigation into S. aureus tolerance mechanisms during food processing and control of biofilm formation.
Collapse
Affiliation(s)
- Ziheng Wu
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xiaolu Lei
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yunying Zhang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Siyao Wu
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Zhiyuan Hou
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Kai Ma
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Hao Pei
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China.
| | - Fei Shang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China.
| | - Ting Xue
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China.
| |
Collapse
|
3
|
Douglas EJ, Palk N, Rudolph ER, Laabei M. Anti-staphylococcal fatty acids: mode of action, bacterial resistance and implications for therapeutic application. MICROBIOLOGY (READING, ENGLAND) 2025; 171:001563. [PMID: 40402078 PMCID: PMC12098983 DOI: 10.1099/mic.0.001563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/29/2025] [Indexed: 05/23/2025]
Abstract
Novel strategies to counter multidrug-resistant pathogens such as methicillin-resistant Staphylococcus aureus are urgently required. The antimicrobial properties of fatty acids (FAs) have long been recognized and offer significant promise as viable alternatives to, or potentiators of, conventional antibiotics. In this review, we examine the interplay between FAs and S. aureus, specifically detailing the underlying molecular mechanisms responsible for FA-mediated inhibition and the counteracting staphylococcal systems evolved to withstand FA onslaught. Finally, we present an update on the recent therapeutic FA applications to combat S. aureus infection, either as a monotherapy or in combination with antibiotics or host-derived antimicrobial peptides. Given the frequency of interaction between FAs and S. aureus during host colonization and infection, understanding FA mode of action and deciphering S. aureus FA resistance strategies are central in rationally designing future anti-staphylococcal FAs and FA-combination therapies.
Collapse
Affiliation(s)
- Edward J.A. Douglas
- Centre for Bacterial Resistance Biology, Imperial College London, London W2 1NY, UK
| | - Nathanael Palk
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - Emily R. Rudolph
- Department of Life Sciences, University of Bath, Bath BA2 7AY, UK
| | - Maisem Laabei
- School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
4
|
Tao Z, Tian C, Zhong C, Ji B, Li W, Zhao Y. The role of NhaA protein in modulating antibiotic tolerance in Escherichia coli. Int J Biol Macromol 2025; 311:143721. [PMID: 40316115 DOI: 10.1016/j.ijbiomac.2025.143721] [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: 12/21/2024] [Revised: 04/08/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
As microbial resistance and recurrent bacterial infections escalate, the growing understanding of the interplay between antibiotic resistance and tolerance has sparked significant interest in the latter. Previous studies have demonstrated that the deletion of cation/proton antiporters (CPAs) induces bacterial phenotypes, such as slow growth and prolonged lag phases, which contribute to the development of tolerance. This study investigates the role of the NhaA protein in antibiotic tolerance in Escherichia coli using CRISPR/Cas9 gene editing to delete the NhaA protein. Our results suggest that the NhaA protein plays a key role in modulating antibiotic tolerance. In response to NhaA deletion, E. coli adapts through multiple mechanisms, including changes in membrane permeability, enhanced efflux activity, increased membrane fluidity, disruption of the proton motive force (PMF), and a reduction in intracellular ATP levels. These adaptive changes collectively promote the development of antibiotic tolerance. Understanding these tolerance mechanisms could uncover new therapeutic targets, help prevent the emergence of tolerance, or sustain bacteria cells in a tolerant state, providing crucial strategies to combat the rise of antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Zhen Tao
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Chuanjun Tian
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Chunyan Zhong
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Bingjie Ji
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Wenwen Li
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China
| | - Yongshan Zhao
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning Province 110016, PR China.
| |
Collapse
|
5
|
Li Z, Lu Y, Song J, Han P, Shi H, Yao X, Zhang X, Zhang G. An emodin-mediated multifunctional nanoplatform with augmented sonodynamic and immunoregulation for osteomyelitis therapy. J Colloid Interface Sci 2025; 684:122-137. [PMID: 39823728 DOI: 10.1016/j.jcis.2025.01.094] [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: 11/17/2024] [Revised: 01/01/2025] [Accepted: 01/12/2025] [Indexed: 01/20/2025]
Abstract
Emodin (ED), as a traditional Chinese medicine, possesses a variety of biological activities and is also one of natural sonosensitizer. Whether emodin could react with titanium dioxide to enhance the sonodynamic activity for safely treating osteomyelitis remains to be explored. Hence, an ED-conjugated Mn-doped titanium dioxide (TOM) nanorod array is designed and prepared on titanium to eliminate bacterial infections under ultrasound (US) treatment. The doping of Mn and the formation of an inorganic-organic interfacial between ED and TOM cause an imbalance in the interfacial charge and reduce the band gap of TOM, thereby increasing the production of reactive oxygen species (ROS) under US. The generated ROS effectively destroys the bacterial cell membrane, accelerating Mn ions intake and thereby inducing ferroptosis-like death of MRSA. Moreover, the hybrid coating activates cAMP and cGMP-PKG pathways to modulate M1 polarization of macrophages to display potent phagocytosis to bacteria. After eliminating the bacterial infection, the coating also can facilitate macrophage M2 polarization to promote osteogenic differentiation. Overall, the Chinese medicine enhanced sonodynamic and immunotherapy have great promise in treating bacterial-infected osteomyelitis.
Collapse
Affiliation(s)
- Zehao Li
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024 China
| | - Ying Lu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032 China; Shanxi Provincial Key Laboratory for Translational Nuclear Medicine and Precision Protection, Taiyuan 030006 China
| | - Jianbo Song
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032 China; Shanxi Provincial Key Laboratory for Translational Nuclear Medicine and Precision Protection, Taiyuan 030006 China
| | - Peide Han
- College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024 China
| | - Huixian Shi
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024 China
| | - Xiaohong Yao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024 China.
| | - Xiangyu Zhang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024 China; College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024 China.
| | - Guannan Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032 China; Shanxi Provincial Key Laboratory for Translational Nuclear Medicine and Precision Protection, Taiyuan 030006 China.
| |
Collapse
|
6
|
Ji Y, Sun C, Wu S. Transcriptomic and Biochemical Analysis of the Antimicrobial Mechanism of Lipopeptide Iturin W against Staphylococcus aureus. Int J Mol Sci 2024; 25:9949. [PMID: 39337437 PMCID: PMC11432370 DOI: 10.3390/ijms25189949] [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: 08/20/2024] [Revised: 09/08/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Staphylococcus aureus is one of the most serious pathogens threatening food safety and public health. We have previously showed that iturin W exhibited obvious antifungal activity on plant pathogens. In the present study, we found iturin W, especially C14 iturin W, showed strong antimicrobial activity against S. aureus, and the antimicrobial mechanism of C14 iturin W was further investigated by transcriptomic analysis and a related biochemical experiment. The results showed that C14 iturin W can reduce the expression levels of genes associated with the reactive oxygen species (ROS) scavenging enzyme and genes involved in arginine biosynthesis, thus leading to the increase in ROS levels of S. aureus. Furthermore, C14 iturin W can also interfere with proton dynamics, which is crucial for cells to regulate various biological possesses. Therefore, ROS accumulation and change in proton motive force are import ways for C14 iturin W to exert the antimicrobial activity. In addition, C14 iturin W can also reduce the expression levels of genes related to virulence factors and decrease the production of enterotoxins and hemolysins in S. aureus, indicating that C14 iturin W has a good potential in food and pharmaceutical fields to reduce the harm caused by S. aureus in the future.
Collapse
Affiliation(s)
- Yingyu Ji
- College of Life Sciences, Qingdao University, Qingdao 266071, China;
| | - Chaomin Sun
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China;
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China
- Center of Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
| | - Shimei Wu
- College of Life Sciences, Qingdao University, Qingdao 266071, China;
| |
Collapse
|
7
|
Wei M, Knight SAB, Fazelinia H, Spruce L, Roof J, Chu E, Kim DY, Bhanap P, Walsh J, Flowers L, Zhu J, Grice EA. An exploration of mechanisms underlying Desemzia incerta colonization resistance to methicillin-resistant Staphylococcus aureus on the skin. mSphere 2024; 9:e0063623. [PMID: 38415632 PMCID: PMC10964421 DOI: 10.1128/msphere.00636-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
Colonization of human skin and nares by methicillin-resistant Staphylococcus aureus (MRSA) leads to the community spread of MRSA. This spread is exacerbated by the transfer of MRSA between humans and livestock, particularly swine. Here, we capitalized on the shared features between human and porcine skin, including shared MRSA colonization, to study novel bacterial mediators of MRSA colonization resistance. We focused on the poorly studied bacterial species Desemzia incerta, which we found to exert antimicrobial activity through a secreted product and exhibited colonization resistance against MRSA in an in vivo murine skin model. Using parallel genomic and biochemical investigation, we discovered that D. incerta secretes an antimicrobial protein. Sequential protein purification and proteomics analysis identified 24 candidate inhibitory proteins, including a promising peptidoglycan hydrolase candidate. Aided by transcriptional analysis of D. incerta and MRSA cocultures, we found that exposure to D. incerta leads to decreased MRSA biofilm production. These results emphasize the value of exploring microbial communities across a spectrum of hosts, which can lead to novel therapeutic agents as well as an increased understanding of microbial competition.IMPORTANCEMethicillin-resistant Staphylococcus aureus (MRSA) causes a significant healthcare burden and can be spread to the human population via livestock transmission. Members of the skin microbiome can prevent MRSA colonization via a poorly understood phenomenon known as colonization resistance. Here, we studied the colonization resistance of S. aureus by bacterial inhibitors previously identified from a porcine skin model. We identify a pig skin commensal, Desemzia incerta, that reduced MRSA colonization in a murine model. We employ a combination of genomic, proteomic, and transcriptomic analyses to explore the mechanisms of inhibition between D. incerta and S. aureus. We identify 24 candidate antimicrobial proteins secreted by D. incerta that could be responsible for its antimicrobial activity. We also find that exposure to D. incerta leads to decreased S. aureus biofilm formation. These findings show that the livestock transmission of MRSA can be exploited to uncover novel mechanisms of MRSA colonization resistance.
Collapse
Affiliation(s)
- Monica Wei
- Department of Dermatology and Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Simon A. B. Knight
- Department of Dermatology and Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Hossein Fazelinia
- Children’s Hospital of Philadelphia, Proteomics Core Facility, Philadelphia, Pennsylvania, USA
| | - Lynn Spruce
- Children’s Hospital of Philadelphia, Proteomics Core Facility, Philadelphia, Pennsylvania, USA
| | - Jennifer Roof
- Children’s Hospital of Philadelphia, Proteomics Core Facility, Philadelphia, Pennsylvania, USA
| | - Emily Chu
- Department of Dermatology and Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Daniel Y. Kim
- Department of Dermatology and Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Preeti Bhanap
- Department of Dermatology and Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jasmine Walsh
- Department of Dermatology and Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Laurice Flowers
- Department of Dermatology and Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jun Zhu
- Department of Dermatology and Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Elizabeth A. Grice
- Department of Dermatology and Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Hilliard JK, Gries CM. Temporal control of Staphylococcus aureus intracellular pH by sodium and potassium. FEMS Microbiol Lett 2024; 371:fnae100. [PMID: 39567841 PMCID: PMC11636269 DOI: 10.1093/femsle/fnae100] [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: 07/26/2024] [Revised: 10/15/2024] [Accepted: 11/19/2024] [Indexed: 11/22/2024] Open
Abstract
Adaptation to environmental change during both colonization and infection is essential to the pathogenesis of Staphylococcus aureus. Like other bacterial pathogens that require potassium to fulfill nutritional and chemiosmotic requirements, S. aureus has been shown to utilize potassium transport to modulate virulence gene expression, antimicrobial resistance, and osmotic tolerance. Recent studies examining the role for potassium uptake in mediating S. aureus physiology have also described its contribution in mediating carbon flux within central metabolism and generation of a proton motive force. Here, we utilize a pH-sensitive green fluorescent protein to examine the temporal regulation of S. aureus intracellular pH by potassium and sodium under various environmental conditions, including extracellular pH and antibiotic stress. Our results distinguish unique conditions and transport mechanisms that utilize these ions to modulate cytoplasmic pH homeostasis, and they identify these processes as a novel mechanism of intrinsic ampicillin resistance in S. aureus.
Collapse
Affiliation(s)
- Julia K Hilliard
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, United States
| | - Casey M Gries
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, United States
| |
Collapse
|
9
|
Beetham CM, Schuster CF, Kviatkovski I, Santiago M, Walker S, Gründling A. Histidine transport is essential for the growth of Staphylococcus aureus at low pH. PLoS Pathog 2024; 20:e1011927. [PMID: 38227607 PMCID: PMC10817146 DOI: 10.1371/journal.ppat.1011927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 01/26/2024] [Accepted: 12/28/2023] [Indexed: 01/18/2024] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen capable of causing many different human diseases. During colonization and infection, S. aureus will encounter a range of hostile environments, including acidic conditions such as those found on the skin and within macrophages. However, little is known about the mechanisms that S. aureus uses to detect and respond to low pH. Here, we employed a transposon sequencing approach to determine on a genome-wide level the genes required or detrimental for growth at low pH. We identified 31 genes that were essential for the growth of S. aureus at pH 4.5 and confirmed the importance of many of them through follow up experiments using mutant strains inactivated for individual genes. Most of the genes identified code for proteins with functions in cell wall assembly and maintenance. These data suggest that the cell wall has a more important role than previously appreciated in promoting bacterial survival when under acid stress. We also identified several novel processes previously not linked to the acid stress response in S. aureus. These include aerobic respiration and histidine transport, the latter by showing that one of the most important genes, SAUSA300_0846, codes for a previously uncharacterized histidine transporter. We further show that under acid stress, the expression of the histidine transporter gene is increased in WT S. aureus. In a S. aureus SAUSA300_0846 mutant strain expression of the histidine biosynthesis genes is induced under acid stress conditions allowing the bacteria to maintain cytosolic histidine levels. This strain is, however, unable to maintain its cytosolic pH to the same extent as a WT strain, revealing an important function specifically for histidine transport in the acid stress response of S. aureus.
Collapse
Affiliation(s)
- Catrin M. Beetham
- Section of Molecular Microbiology and Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Christopher F. Schuster
- Section of Molecular Microbiology and Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Igor Kviatkovski
- Section of Molecular Microbiology and Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Marina Santiago
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Suzanne Walker
- Department of Microbiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Angelika Gründling
- Section of Molecular Microbiology and Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| |
Collapse
|
10
|
Hong YH, Huang HM, Wu L, Storey KB, Zhang JY, Zhang YP, Yu DN. Characterization of Two Mitogenomes of Hyla sanchiangensis (Anura: Hylidae), with Phylogenetic Relationships and Selection Pressure Analyses of Hylidae. Animals (Basel) 2023; 13:ani13101593. [PMID: 37238023 DOI: 10.3390/ani13101593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Hyla sanchiangensis (Anura: Hylidae) is endemic to China and is distributed across Anhui, Zhejiang, Fujian, Guangdong, Guangxi, Hunan, and Guizhou provinces. The mitogenomes of H. sanchiangensis from two different sites (Jinxiu, Guangxi, and Wencheng, Zhejiang) were sequenced. Phylogenetic analyses were conducted, including 38 mitogenomes of Hylidae from the NCBI database, and assessed the phylogenetic relationship of H. sanchiangensis within the analyzed dataset. Two mitogenomes of H. sanchiangensis showed the typical mitochondrial gene arrangement with 13 protein-coding genes (PCGs), two ribosomal RNA genes (12S rRNA and 16S rRNA), 22 transfer RNA (tRNA) genes, and one non-coding control region (D-loop). The lengths of the 12S rRNA and 16S rRNA genes from both samples (Jinxiu and Wencheng) were 933 bp and 1604 bp, respectively. The genetic distance (p-distance transformed into percent) on the basis of the mitogenomes (excluding the control region) of the two samples was calculated as 4.4%. Hyla sanchiangensis showed a close phylogenetic relationship with the clade of (H. annectans + H. tsinlingensis), which was supported by ML and BI analyses. In the branch-site model, five positive selection sites were found in the clade of Hyla and Dryophytes: Cytb protein (at position 316), ND3 protein (at position 85), and ND5 protein (at position 400) have one site, respectively, and two sites in ND4 protein (at positions 47 and 200). Based on the results, we hypothesized that the positive selection of Hyla and Dryophytes was due to their experience of cold stress in historical events, but more evidence is needed to support this conclusion.
Collapse
Affiliation(s)
- Yue-Huan Hong
- College of Life Sciences, Zhejiang Normal University, Jinhua 321004, China
| | | | - Lian Wu
- College of Life Sciences, Zhejiang Normal University, Jinhua 321004, China
| | - Kenneth B Storey
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada
| | - Jia-Yong Zhang
- College of Life Sciences, Zhejiang Normal University, Jinhua 321004, China
- Key Lab of Wildlife Biotechnology, Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, Jinhua 321004, China
| | - Yong-Pu Zhang
- College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Dan-Na Yu
- College of Life Sciences, Zhejiang Normal University, Jinhua 321004, China
- Key Lab of Wildlife Biotechnology, Conservation and Utilization of Zhejiang Province, Zhejiang Normal University, Jinhua 321004, China
| |
Collapse
|
11
|
Wicaksono WA, Egamberdieva D, Berg C, Mora M, Kusstatscher P, Cernava T, Berg G. Function-Based Rhizosphere Assembly along a Gradient of Desiccation in the Former Aral Sea. mSystems 2022; 7:e0073922. [PMID: 36377901 PMCID: PMC9765073 DOI: 10.1128/msystems.00739-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/29/2022] [Indexed: 11/16/2022] Open
Abstract
The desiccation of the Aral Sea represents one of the largest human-made environmental regional disasters. The salt- and toxin-enriched dried-out basin provides a natural laboratory for studying ecosystem functioning and rhizosphere assembly under extreme anthropogenic conditions. Here, we investigated the prokaryotic rhizosphere communities of the native pioneer plant Suaeda acuminata (C.A.Mey.) Moq. in comparison to bulk soil across a gradient of desiccation (5, 10, and 40 years) by metagenome and amplicon sequencing combined with quantitative PCR (qPCR) analyses. The rhizosphere effect was evident due to significantly higher bacterial abundances but less diversity in the rhizosphere compared to bulk soil. Interestingly, in the highest salinity (5 years of desiccation), rhizosphere functions were mainly provided by archaeal communities. Along the desiccation gradient, we observed a significant change in the rhizosphere microbiota, which was reflected by (i) a decreasing archaeon-bacterium ratio, (ii) replacement of halophilic archaea by specific plant-associated bacteria, i.e., Alphaproteobacteria and Actinobacteria, and (iii) an adaptation of specific, potentially plant-beneficial biosynthetic pathways. In general, both bacteria and archaea were found to be involved in carbon cycling and fixation, as well as methane and nitrogen metabolism. Analysis of metagenome-assembled genomes (MAGs) showed specific signatures for production of osmoprotectants, assimilatory nitrate reduction, and transport system induction. Our results provide evidence that rhizosphere assembly by cofiltering specific taxa with distinct traits is a mechanism which allows plants to thrive under extreme conditions. Overall, our findings highlight a function-based rhizosphere assembly, the importance of plant-microbe interactions in salinated soils, and their exploitation potential for ecosystem restoration approaches. IMPORTANCE The desertification of the Aral Sea basin in Uzbekistan and Kazakhstan represents one of the most serious anthropogenic environmental disasters of the last century. Since the 1960s, the world's fourth-largest inland body of water has been constantly shrinking, which has resulted in an extreme increase of salinity accompanied by accumulation of many hazardous and carcinogenic substances, as well as heavy metals, in the dried-out basin. Here, we investigated bacterial and archaeal communities in the rhizosphere of pioneer plants by combining classic molecular methods with amplicon sequencing as well as metagenomics for functional insights. By implementing a desiccation gradient, we observed (i) remarkable differences in the archaeon-bacterium ratio of plant rhizosphere samples, (ii) replacement of archaeal indicator taxa during succession, and (iii) the presence of specific, potentially plant-beneficial biosynthetic pathways in archaea present during the early stages. In addition, our results provide hitherto-undescribed insights into the functional redundancy between plant-associated archaea and bacteria.
Collapse
Affiliation(s)
- Wisnu Adi Wicaksono
- Institute of Environmental Biotechnology, Graz University of Technology, Graz, Austria
| | | | | | - Maximilian Mora
- Institute of Environmental Biotechnology, Graz University of Technology, Graz, Austria
| | - Peter Kusstatscher
- Institute of Environmental Biotechnology, Graz University of Technology, Graz, Austria
| | - Tomislav Cernava
- Institute of Environmental Biotechnology, Graz University of Technology, Graz, Austria
| | - Gabriele Berg
- Institute of Environmental Biotechnology, Graz University of Technology, Graz, Austria
- Leibniz Institute for Agricultural Engineering and Bioeconomy, Potsdam, Germany
- Institute for Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| |
Collapse
|
12
|
Hubloher JJ, van der Sande L, Müller V. Na + homeostasis in Acinetobacter baumannii is facilitated via the activity of the Mrp antiporter. Environ Microbiol 2022; 24:4411-4424. [PMID: 35535800 DOI: 10.1111/1462-2920.16039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/26/2022] [Accepted: 05/04/2022] [Indexed: 11/30/2022]
Abstract
The human opportunistic pathogen Acinetobacter baumannii is a global threat to healthcare institutions worldwide, since it developed very efficient strategies to evade host defense and to adapt to the different environmental conditions of the host. This worked focused on the importance of Na+ homeostasis in A. baumannii with regards to pathobiological aspects. In silico studies revealed a homologue of a multicomponent Na+ /H+ antiporter system. Inactivation of the Mrp antiporter through deletion of the first gene (mrpA') resulted in a mutant that was sensitive to increasing pH values. Furthermore, the strain was highly sensitive to increasing Na+ and Li+ concentrations. Increasing Na+ sensitivity is thought to be responsible for growth impairment in human fluids. Furthermore, deletion of mrpA' is associated with energetic defects, inhibition of motility and survival under anoxic and dry conditions.
Collapse
Affiliation(s)
- Josephine Joy Hubloher
- Department of Molecular Microbiology & Bioenergetics, Institute of Molecular Biosciences, Goethe-University Frankfurt am Main, Germany
| | - Lisa van der Sande
- Department of Molecular Microbiology & Bioenergetics, Institute of Molecular Biosciences, Goethe-University Frankfurt am Main, Germany
| | - Volker Müller
- Department of Molecular Microbiology & Bioenergetics, Institute of Molecular Biosciences, Goethe-University Frankfurt am Main, Germany
| |
Collapse
|
13
|
Tang H, Qu X, Zhang W, Chen X, Zhang S, Xu Y, Yang H, Wang Y, Yang J, Yuan WE, Yue B. Photosensitizer Nanodot Eliciting Immunogenicity for Photo-Immunologic Therapy of Postoperative Methicillin-Resistant Staphylococcus aureus Infection and Secondary Recurrence. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107300. [PMID: 34865257 DOI: 10.1002/adma.202107300] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/11/2021] [Indexed: 06/13/2023]
Abstract
The treatment of postoperative infection caused by multidrug-resistant bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA), has become an intractable clinical challenge owing to its low therapeutic efficacy and high risk of recurrence. Apart from imperfect antibacterial therapies, induction of insufficient immunogenicity, required for the successful clearance of a pathogen, may also contribute to the problem. Herein, an ultra-micro photosensitizer, AgB nanodots, using photothermal therapy, photodynamic therapy, and Ag+ ion sterilization, are utilized to efficiently clear invading MRSA both in vitro and in vivo. AgB nanodots are also found to upregulate host immunogenicity in a murine model and establish immunological memory by promoting the upregulated expression of danger signals that are commonly induced by stress-related responses, including sudden temperature spikes or excess reactive oxygen production. These stimulations boost the antibacterial effects of macrophages, dendritic cells, T cells, or even memory B cells, which is usually defined as infection-related immunogenic cell death. Hence, the proposed AgB nanodot strategy may offer a novel platform for the effective treatment of postoperative infection while providing a systematic immunotherapeutic strategy to combat persistent infections, thereby markedly reducing the incidence of recurrence following recovery from primary infections.
Collapse
Affiliation(s)
- Haozheng Tang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shandong Middle Road, Shanghai, 200001, P. R. China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shandong Middle Road, Shanghai, 200001, P. R. China
| | - Wenkai Zhang
- Pharm-X Center, Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Xuan Chen
- Pharm-X Center, Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Shutao Zhang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shandong Middle Road, Shanghai, 200001, P. R. China
| | - Yang Xu
- Pharm-X Center, Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Hongtao Yang
- Department of Plastic and Reconstructive Surgery, The Ohio State University, Columbus, OH, 43210, USA
- School of Medical Science and Engineering, Beihang University, Beijing, 100191, P. R. China
| | - You Wang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shandong Middle Road, Shanghai, 200001, P. R. China
| | - Jianping Yang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Wei-En Yuan
- Pharm-X Center, Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Bing Yue
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 145 Shandong Middle Road, Shanghai, 200001, P. R. China
| |
Collapse
|
14
|
Transcriptome Analysis of Halotolerant Staphylococcus saprophyticus Isolated from Korean Fermented Shrimp. Foods 2022; 11:foods11040524. [PMID: 35206000 PMCID: PMC8870806 DOI: 10.3390/foods11040524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 11/16/2022] Open
Abstract
Saeu-jeotgal, a Korean fermented shrimp food, is commonly used as an ingredient for making kimchi and other side dishes. The high salinity of the jeotgal contributes to its flavor and inhibits the growth of food spoilage microorganisms. Interestingly, Staphylococcus saprophyticus was discovered to be capable of growth even after treatment with 20% NaCl. To elucidate the tolerance mechanism, a genome-wide gene expression of S. saprophyticus against 0%, 10%, and 20% NaCl was investigated by RNA sequencing. A total of 831, 1314, and 1028 differentially expressed genes (DEGs) were identified in the 0% vs. 10%, 0% vs. 20%, and 10% vs. 20% NaCl comparisons, respectively. The Clusters of Orthologous Groups analysis revealed that the DEGs were involved in amino acid transport and metabolism, transcription, and inorganic ion transport and metabolism. The functional enrichment analysis showed that the expression of the genes encoding mechanosensitive ion channels, sodium/proton antiporters, and betaine/carnitine/choline transporter family proteins was downregulated, whereas the expression of the genes encoding universal stress proteins and enzymes for glutamate, glycine, and alanine synthesis was upregulated. Therefore, these findings suggest that the S. saprophyticus isolated from the saeu-jeotgal utilizes different molecular strategies for halotolerance, with glutamate as the key molecule.
Collapse
|
15
|
Hreha TN, Foreman S, Duran-Pinedo A, Morris AR, Diaz-Rodriguez P, Jones JA, Ferrara K, Bourges A, Rodriguez L, Koffas MAG, Hahn M, Hauser AR, Barquera B. The three NADH dehydrogenases of Pseudomonas aeruginosa: Their roles in energy metabolism and links to virulence. PLoS One 2021; 16:e0244142. [PMID: 33534802 PMCID: PMC7857637 DOI: 10.1371/journal.pone.0244142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 12/03/2020] [Indexed: 11/19/2022] Open
Abstract
Pseudomonas aeruginosa is a ubiquitous opportunistic pathogen which relies on a highly adaptable metabolism to achieve broad pathogenesis. In one example of this flexibility, to catalyze the NADH:quinone oxidoreductase step of the respiratory chain, P. aeruginosa has three different enzymes: NUO, NQR and NDH2, all of which carry out the same redox function but have different energy conservation and ion transport properties. In order to better understand the roles of these enzymes, we constructed two series of mutants: (i) three single deletion mutants, each of which lacks one NADH dehydrogenase and (ii) three double deletion mutants, each of which retains only one of the three enzymes. All of the mutants grew approximately as well as wild type, when tested in rich and minimal medium and in a range of pH and [Na+] conditions, except that the strain with only NUO (ΔnqrFΔndh) has an extended lag phase. During exponential phase, the NADH dehydrogenases contribute to total wild-type activity in the following order: NQR > NDH2 > NUO. Some mutants, including the strain without NQR (ΔnqrF) had increased biofilm formation, pyocyanin production, and killed more efficiently in both macrophage and mouse infection models. Consistent with this, ΔnqrF showed increased transcription of genes involved in pyocyanin production.
Collapse
Affiliation(s)
- Teri N. Hreha
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Sara Foreman
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Ana Duran-Pinedo
- Department of Oral Biology, University of Florida, College of Dentistry, Gainesville, Florida, United States of America
| | - Andrew R. Morris
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Patricia Diaz-Rodriguez
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - J. Andrew Jones
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Kristina Ferrara
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Anais Bourges
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Lauren Rodriguez
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Mattheos A. G. Koffas
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Mariah Hahn
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Alan R. Hauser
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Blanca Barquera
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- * E-mail:
| |
Collapse
|
16
|
Casey D, Sleator RD. A genomic analysis of osmotolerance in Staphylococcus aureus. Gene 2020; 767:145268. [PMID: 33157201 DOI: 10.1016/j.gene.2020.145268] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/07/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022]
Abstract
A key phenotypic characteristic of the Gram-positive bacterial pathogen, Staphylococcus aureus, is its ability to grow in low aw environments. A homology transfer based approach, using the well characterised osmotic stress response systems of Bacillus subtilis and Escherichia coli, was used to identify putative osmotolerance loci in Staphylococcus aureus ST772-MRSA-V. A total of 17 distinct putative hyper and hypo-osmotic stress response systems, comprising 78 genes, were identified. The ST772-MRSA-V genome exhibits significant degeneracy in terms of the osmotic stress response; with three copies of opuD, two copies each of nhaK and mrp/mnh, and five copies of opp. Furthermore, regulation of osmotolerance in ST772-MRSA-V appears to be mediated at the transcriptional, translational, and post-translational levels.
Collapse
Affiliation(s)
- Dylan Casey
- Department of Biological Sciences, Munster Technological University, Bishopstown Campus, Cork, Ireland
| | - Roy D Sleator
- Department of Biological Sciences, Munster Technological University, Bishopstown Campus, Cork, Ireland.
| |
Collapse
|
17
|
Chu J, Koirala B, Forelli N, Vila-Farres X, Ternei MA, Ali T, Colosimo DA, Brady SF. Synthetic-Bioinformatic Natural Product Antibiotics with Diverse Modes of Action. J Am Chem Soc 2020; 142:14158-14168. [PMID: 32697091 DOI: 10.1021/jacs.0c04376] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bacterial natural products have inspired the development of numerous antibiotics in use today. As resistance to existing antibiotics has become more prevalent, new antibiotic lead structures and activities are desperately needed. An increasing number of natural product biosynthetic gene clusters, to which no known molecules can be assigned, are found in genome and metagenome sequencing data. Here we access structural information encoded in this underexploited resource using a synthetic-bioinformatic natural product (syn-BNP) approach, which relies on bioinformatic algorithms followed by chemical synthesis to predict and then produce small molecules inspired by biosynthetic gene clusters. In total, 157 syn-BNP cyclic peptides inspired by 96 nonribosomal peptide synthetase gene clusters were synthesized and screened for antibacterial activity. This yielded nine antibiotics with activities against ESKAPE pathogens as well as Mycobacterium tuberculosis. Not only are antibiotic-resistant pathogens susceptible to many of these syn-BNP antibiotics, but they were also unable to develop resistance to these antibiotics in laboratory experiments. Characterized modes of action for these antibiotics include cell lysis, membrane depolarization, inhibition of cell wall biosynthesis, and ClpP protease dysregulation. Increasingly refined syn-BNP-based explorations of biosynthetic gene clusters should allow for more rapid identification of evolutionarily inspired bioactive small molecules, in particular antibiotics with diverse mechanism of actions that could help confront the imminent crisis of antimicrobial resistance.
Collapse
Affiliation(s)
- John Chu
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Bimal Koirala
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Nicholas Forelli
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Xavier Vila-Farres
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Melinda A Ternei
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Thahmina Ali
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Dominic A Colosimo
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| | - Sean F Brady
- Laboratory of Genetically Encoded Small Molecules, The Rockefeller University, 1230 York Avenue, New York, New York 10065, United States
| |
Collapse
|
18
|
Putative Horizontally Acquired Genes, Highly Transcribed during Yersinia pestis Flea Infection, Are Induced by Hyperosmotic Stress and Function in Aromatic Amino Acid Metabolism. J Bacteriol 2020; 202:JB.00733-19. [PMID: 32205462 PMCID: PMC7221256 DOI: 10.1128/jb.00733-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/17/2020] [Indexed: 11/20/2022] Open
Abstract
Distinct gene repertoires are expressed during Y. pestis infection of its flea and mammalian hosts. The functions of many of these genes remain predicted or unknown, necessitating their characterization, as this may provide a better understanding of Y. pestis specialized biological adaptations to the discrete environments of its two hosts. This study provides functional context to adjacently clustered horizontally acquired genes predominantly expressed in the flea host by deciphering their fundamental processes with regard to (i) transcriptional organization, (ii) transcription activation signals, and (iii) biochemical function. Our data support a role for these genes in osmoadaptation and aromatic amino acid metabolism, highlighting these as preferential processes by which Y. pestis gene expression is modulated during flea infection. While alternating between insects and mammals during its life cycle, Yersinia pestis, the flea-transmitted bacterium that causes plague, regulates its gene expression appropriately to adapt to these two physiologically disparate host environments. In fleas competent to transmit Y. pestis, low-GC-content genes y3555, y3551, and y3550 are highly transcribed, suggesting that these genes have a highly prioritized role in flea infection. Here, we demonstrate that y3555, y3551, and y3550 are transcribed as part of a single polycistronic mRNA comprising the y3555, y3554, y3553, y355x, y3551, and y3550 genes. Additionally, y355x-y3551-y3550 compose another operon, while y3550 can be also transcribed as a monocistronic mRNA. The expression of these genes is induced by hyperosmotic salinity stress, which serves as an explicit environmental stimulus that initiates transcriptional activity from the predicted y3550 promoter. Y3555 has homology to pyridoxal 5′-phosphate (PLP)-dependent aromatic aminotransferases, while Y3550 and Y3551 are homologous to the Rid protein superfamily (YjgF/YER057c/UK114) members that forestall damage caused by reactive intermediates formed during PLP-dependent enzymatic activity. We demonstrate that y3551 specifically encodes an archetypal RidA protein with 2-aminoacrylate deaminase activity but Y3550 lacks Rid deaminase function. Heterologous expression of y3555 generates a critical aspartate requirement in a Salmonella entericaaspC mutant, while its in vitro expression, and specifically its heterologous coexpression with y3550, enhances the growth rate of an Escherichia coli ΔaspC ΔtyrB mutant in a defined minimal amino acid-supplemented medium. Our data suggest that the y3555, y3551, and y3550 genes operate cooperatively to optimize aromatic amino acid metabolism and are induced under conditions of hyperosmotic salinity stress. IMPORTANCE Distinct gene repertoires are expressed during Y. pestis infection of its flea and mammalian hosts. The functions of many of these genes remain predicted or unknown, necessitating their characterization, as this may provide a better understanding of Y. pestis specialized biological adaptations to the discrete environments of its two hosts. This study provides functional context to adjacently clustered horizontally acquired genes predominantly expressed in the flea host by deciphering their fundamental processes with regard to (i) transcriptional organization, (ii) transcription activation signals, and (iii) biochemical function. Our data support a role for these genes in osmoadaptation and aromatic amino acid metabolism, highlighting these as preferential processes by which Y. pestis gene expression is modulated during flea infection.
Collapse
|
19
|
Schuster CF, Wiedemann DM, Kirsebom FCM, Santiago M, Walker S, Gründling A. High-throughput transposon sequencing highlights the cell wall as an important barrier for osmotic stress in methicillin resistant Staphylococcus aureus and underlines a tailored response to different osmotic stressors. Mol Microbiol 2019; 113:699-717. [PMID: 31770461 PMCID: PMC7176532 DOI: 10.1111/mmi.14433] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 11/24/2019] [Indexed: 12/28/2022]
Abstract
Staphylococcus aureus is an opportunistic pathogen that can cause soft tissue infections but is also a frequent cause of foodborne illnesses. One contributing factor for this food association is its high salt tolerance allowing this organism to survive commonly used food preservation methods. How this resistance is mediated is poorly understood, particularly during long-term exposure. In this study, we used transposon sequencing (TN-seq) to understand how the responses to osmotic stressors differ. Our results revealed distinctly different long-term responses to NaCl, KCl and sucrose stresses. In addition, we identified the DUF2538 domain containing gene SAUSA300_0957 (gene 957) as essential under salt stress. Interestingly, a 957 mutant was less susceptible to oxacillin and showed increased peptidoglycan crosslinking. The salt sensitivity phenotype could be suppressed by amino acid substitutions in the transglycosylase domain of the penicillin-binding protein Pbp2, and these changes restored the peptidoglycan crosslinking to WT levels. These results indicate that increased crosslinking of the peptidoglycan polymer can be detrimental and highlight a critical role of the bacterial cell wall for osmotic stress resistance. This study will serve as a starting point for future research on osmotic stress response and help develop better strategies to tackle foodborne staphylococcal infections.
Collapse
Affiliation(s)
- Christopher F Schuster
- Section of Molecular Microbiology and MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - David M Wiedemann
- Section of Molecular Microbiology and MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Freja C M Kirsebom
- Section of Molecular Microbiology and MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Marina Santiago
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Suzanne Walker
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Angelika Gründling
- Section of Molecular Microbiology and MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| |
Collapse
|
20
|
Nguyen T, Kim T, Ta HM, Yeo WS, Choi J, Mizar P, Lee SS, Bae T, Chaurasia AK, Kim KK. Targeting Mannitol Metabolism as an Alternative Antimicrobial Strategy Based on the Structure-Function Study of Mannitol-1-Phosphate Dehydrogenase in Staphylococcus aureus. mBio 2019; 10:e02660-18. [PMID: 31289190 PMCID: PMC6623548 DOI: 10.1128/mbio.02660-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 06/05/2019] [Indexed: 11/22/2022] Open
Abstract
Mannitol-1-phosphate dehydrogenase (M1PDH) is a key enzyme in Staphylococcus aureus mannitol metabolism, but its roles in pathophysiological settings have not been established. We performed comprehensive structure-function analysis of M1PDH from S. aureus USA300, a strain of community-associated methicillin-resistant S. aureus, to evaluate its roles in cell viability and virulence under pathophysiological conditions. On the basis of our results, we propose M1PDH as a potential antibacterial target. In vitro cell viability assessment of ΔmtlD knockout and complemented strains confirmed that M1PDH is essential to endure pH, high-salt, and oxidative stress and thus that M1PDH is required for preventing osmotic burst by regulating pressure potential imposed by mannitol. The mouse infection model also verified that M1PDH is essential for bacterial survival during infection. To further support the use of M1PDH as an antibacterial target, we identified dihydrocelastrol (DHCL) as a competitive inhibitor of S. aureus M1PDH (SaM1PDH) and confirmed that DHCL effectively reduces bacterial cell viability during host infection. To explain physiological functions of SaM1PDH at the atomic level, the crystal structure of SaM1PDH was determined at 1.7-Å resolution. Structure-based mutation analyses and DHCL molecular docking to the SaM1PDH active site followed by functional assay identified key residues in the active site and provided the action mechanism of DHCL. Collectively, we propose SaM1PDH as a target for antibiotic development based on its physiological roles with the goals of expanding the repertory of antibiotic targets to fight antimicrobial resistance and providing essential knowledge for developing potent inhibitors of SaM1PDH based on structure-function studies.IMPORTANCE Due to the shortage of effective antibiotics against drug-resistant Staphylococcus aureus, new targets are urgently required to develop next-generation antibiotics. We investigated mannitol-1-phosphate dehydrogenase of S. aureus USA300 (SaM1PDH), a key enzyme regulating intracellular mannitol levels, and explored the possibility of using SaM1PDH as a target for developing antibiotic. Since mannitol is necessary for maintaining the cellular redox and osmotic potential, the homeostatic imbalance caused by treatment with a SaM1PDH inhibitor or knockout of the gene encoding SaM1PDH results in bacterial cell death through oxidative and/or mannitol-dependent cytolysis. We elucidated the molecular mechanism of SaM1PDH and the structural basis of substrate and inhibitor recognition by enzymatic and structural analyses of SaM1PDH. Our results strongly support the concept that targeting of SaM1PDH represents an alternative strategy for developing a new class of antibiotics that cause bacterial cell death not by blocking key cellular machinery but by inducing cytolysis and reducing stress tolerance through inhibition of the mannitol pathway.
Collapse
Affiliation(s)
- Thanh Nguyen
- Department of Molecular Cell Biology, Institute for Antimicrobial Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Truc Kim
- Department of Molecular Cell Biology, Institute for Antimicrobial Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Hai Minh Ta
- Department of Molecular Cell Biology, Institute for Antimicrobial Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Won Sik Yeo
- Department of Microbiology and Immunology, Indiana University School of Medicine Northwest, Gary, Indiana, USA
| | - Jongkeun Choi
- Department of Chemical Engineering, Chungwoon University, Incheon, South Korea
| | - Pushpak Mizar
- School of Chemistry, University of Southampton, Southampton, United Kingdom
| | - Seung Seo Lee
- School of Chemistry, University of Southampton, Southampton, United Kingdom
| | - Taeok Bae
- Department of Microbiology and Immunology, Indiana University School of Medicine Northwest, Gary, Indiana, USA
| | - Akhilesh Kumar Chaurasia
- Department of Molecular Cell Biology, Institute for Antimicrobial Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Kyeong Kyu Kim
- Department of Molecular Cell Biology, Institute for Antimicrobial Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, South Korea
- Samsung Biomedical Research Institute, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
21
|
Artificial Selection for Pathogenicity Mutations in Staphylococcus aureus Identifies Novel Factors Relevant to Chronic Infection. Infect Immun 2019; 87:IAI.00884-18. [PMID: 30642903 DOI: 10.1128/iai.00884-18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 01/10/2019] [Indexed: 02/06/2023] Open
Abstract
Adaptation of Staphylococcus aureus to host microenvironments during chronic infection involves spontaneous mutations, yet changes underlying adaptive phenotypes remain incompletely explored. Here, we employed artificial selection and whole-genome sequencing to better characterize spontaneous chromosomal mutations that alter two pathogenicity phenotypes relevant to chronic infection in S. aureus: intracellular invasiveness and intracellular cytotoxicity. We identified 23 genes whose alteration coincided with enhanced virulence, 11 that were previously known and 12 (52%) that had no previously described role in S. aureus pathogenicity. Using precision genome editing, transposon mutants, and gene complementation, we empirically assessed the contributions of individual genes to the two virulence phenotypes. We functionally validated 14 of 21 genes tested as measurably influencing invasion and/or cytotoxicity, including 8 newly implicated by this study. We identified inactivating mutations (murA, ndhC, and a hypothetical membrane protein) and gain-of-function mutations (aroE Thr182Ile, yhcF Thr74Ile, and Asp486Glu in a hypothetical peptidase) in previously unrecognized S. aureus virulence genes that enhance pathogenesis when introduced into a clean genetic background, as well as a novel activating mutation in the known virulence regulator gene saeS (Ala106Thr). Investigation of potentially epistatic interactions identified a tufA mutation (Ala271Val) that enhances virulence only in the context of purine operon repressor gene (purR) inactivation. This project reveals a functionally diverse range of genes affected by gain- or loss-of-function mutations that contribute to S. aureus adaptive virulence phenotypes. More generally, the work establishes artificial selection as a means to determine the genetic mechanisms underlying complex bacterial phenotypes relevant to adaptation during infection.
Collapse
|