1
|
Jeon S, Baek H, Kim S, Kim Y, Kim J, Kim JW. Microalgae-Derived Microparticles Improve Immunomodulation via Combined Glycolysis and MAPK Activation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:8619-8626. [PMID: 40145572 DOI: 10.1021/acs.langmuir.4c05088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Natural polysaccharides possess potent immunostimulatory properties, but their poor solubility impedes efficiency of cellular delivery. This study focuses on extraction of microparticles (MPs) fromEuglena gracilis, a microalgae species characterized by abundant intracellular β-1,3-glucan and flexible cell membrane. We introduce anE. gracilis-derived MP (MPEG) system as a natural carrier for solubilized β-glucan. The MPEG system enhances β-glucan's solubility and loading efficiency through sequential sonication and cell extrusion. In vitro studies reveal that MPEG utilizes multiple endocytosis pathways, including phagocytosis, clathrin-mediated, and lipid raft-mediated routes, for effective β-glucan delivery into cells. Upon cellular internalization, MPEG components trigger dual activation of the MAPK signaling pathway and glycolysis in macrophages, leading to enhanced production of pro-inflammatory cytokines and lactic acid, ultimately strengthening innate immune responses. This MPEG system offers a promising approach to harnessing the immunostimulatory properties of natural polysaccharides while overcoming their solubility limitations, opening new avenues for targeted cellular delivery in immunomodulation therapies.
Collapse
Affiliation(s)
- Saetbyeol Jeon
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hwira Baek
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seulgi Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Youngseok Kim
- Technology Innovation Center, Shinsegae International Inc., Seoul 06015, Republic of Korea
| | - Junoh Kim
- Technology Innovation Center, Shinsegae International Inc., Seoul 06015, Republic of Korea
| | - Jin Woong Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
2
|
Pokorzynski ND, Jones KA, Campagna SR, Groisman EA. Cytoplasmic Mg 2+ supersedes carbon source preference to dictate Salmonella metabolism. Proc Natl Acad Sci U S A 2025; 122:e2424337122. [PMID: 40131949 PMCID: PMC12002343 DOI: 10.1073/pnas.2424337122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Glucose is the preferred carbon source of most studied microorganisms. However, we now report that glucose loses preferred status when the intracellular pathogen Salmonella enterica serovar Typhimurium experiences cytoplasmic magnesium (Mg2+) starvation. We establish that this infection-relevant stress drastically reduces synthesis of cyclic adenosine monophosphate (cAMP), the allosteric activator of the cAMP receptor protein (CRP), master regulator of carbon utilization. The resulting reduction in cAMP concentration, which is independent of carbon source, decreases transcription of CRP-cAMP-activated carbon utilization genes, hinders carbon source uptake, and restricts metabolism, rendering wild-type bacteria phenotypically CRP-. A cAMP-independent allele of CRP overcame the transcriptional, uptake, and metabolic restrictions caused by cytoplasmic Mg2+ starvation and significantly increased transcription of the glucose uptake gene when S. Typhimurium was inside murine macrophages. The reduced preference for glucose exhibited by S. Typhimurium inside macrophages reflects that transcription of the glucose uptake gene requires higher amounts of active CRP-cAMP than transcription of uptake genes for preferred carbon sources, such as gluconate and glycerol. By reducing CRP-cAMP activity, low cytoplasmic Mg2+ alters carbon source preference, adjusting metabolism and growth.
Collapse
Affiliation(s)
- Nick D. Pokorzynski
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT06536
| | - Katarina A. Jones
- Biological and Small Molecule Mass Spectrometry Core, University of Tennessee, Knoxville, TN37996
| | - Shawn R. Campagna
- Biological and Small Molecule Mass Spectrometry Core, University of Tennessee, Knoxville, TN37996
| | - Eduardo A. Groisman
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT06536
| |
Collapse
|
3
|
Wu A, Liang C, Chen W, Lu C, Chen J, Wu B, Chen D, He L, Wang X. ZnO-Cu/Mn nanozyme for rescuing the intestinal homeostasis in Salmonella-induced colitis. J Nanobiotechnology 2025; 23:225. [PMID: 40114178 PMCID: PMC11924796 DOI: 10.1186/s12951-025-03283-4] [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: 01/22/2025] [Accepted: 02/28/2025] [Indexed: 03/22/2025] Open
Abstract
Salmonella is one of the most common foodborne pathogens, which can cause severe enteritis and intestinal microbiota imbalance. However, there are limited strategies currently available for preventing or treating Salmonella-induced colitis. Herein, we developed the Cu/Mn-co-doped ZnO tandem nanozyme (ZnO-CM) with pH-responsive multienzyme-mimicking activities via doping engineering for the treatment of Salmonella-induced colitis. Benefiting from the co-doping of Cu and Mn, ZnO-CM nanospheres exhibit remarkable peroxidase-like activity in acidic condition and superoxide dismutase- and catalase-like activities in neutral environment. Animal experiments show that ZnO-CM can efficiently inhibit bacterial growth, alleviate inflammation, and restore the intestinal barrier, resulting in good antibacterial and anti-inflammatory effects on Salmonella-induced colitis. Mechanistically, ZnO-CM functions through inhibiting the continuous accumulation of ROS, increasing the levels of tight junction proteins occludin and claudin-1, and decreasing the expression of pro-inflammatory cytokines IL-1β and IL-6 in intestine. This work not only presents an effective paradigm for Salmonella-induced colitis therapy, but also provides new sights into the prevention and treatment of other bacterial enteritis.
Collapse
Affiliation(s)
- Aimin Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, China
| | - Chen Liang
- College of Science, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - WenShuang Chen
- College of Science, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - ChangFang Lu
- College of Science, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - JunZhou Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, China
| | - Bing Wu
- Sichuan Chelota Biotechnology Group Co., Ltd, Chengdu, 618302, Sichuan, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Li He
- College of Science, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| | - Xianxiang Wang
- College of Science, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
4
|
Zhou L, Cai X, Dong Q, Yin J, Liu Y, Gao X, Jiang Q, Zhang Y, Zhang X. The immune response and autophagy of Macrobrachium rosenbergii against Aeromonas veronii infection. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101488. [PMID: 40112606 DOI: 10.1016/j.cbd.2025.101488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/20/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
Aeromonas is a bacterial pathogen that causes significant economic losses in the Macrobrachium rosenbergii industry. This study evaluated the transcriptome analysis of M. rosenbergii infected with A. veronii and examined the gene expression patterns associated with immunity in the gills, muscles, intestines, and hepatopancreas. Specifically, 47,988 unigenes and 15,604 differentially expressed genes (DEGs) were identified. The immune-related DEGs were primarily enriched in 20 innate immune signaling pathways, including the NOD-like receptor, Toll-like receptor, and RIG-I-like receptor signaling pathways, etc., as determined by KEGG enrichment analysis. Notably, autophagy-related genes ATG5, ATG12, ATG16L1, and ATG8 were enriched in the NOD-like receptor signaling pathways. Moreover, ATG12, ATG16L1, and ATG8 exhibited significantly up-regulated expression to varying degrees in the hepatopancreas, gills, muscles, and intestines at 12, 24, 36, and 48 h post-infection (hpi). In addition, many autolysosomes were observed in hepatopancreas cells of infected prawns using transmission electron microscopy (TEM). Ultimately, we identified the autophagy-related genes implicated in the immune response of M. rosenbergii, offering a theoretical foundation for elucidating the role of autophagy in the prawn's innate immune mechanisms.
Collapse
Affiliation(s)
- Liying Zhou
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Xiaoyu Cai
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Qi Dong
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Jia Yin
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yan Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Xiaojian Gao
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Qun Jiang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yao Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Xiaojun Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
5
|
Arias AM, Reinartz DM, Sairs C, Kumar SS, Wilson JE. Streptococcus anginosus Activates the NLRP3 Inflammasome to Promote Inflammatory Responses from Macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642696. [PMID: 40161672 PMCID: PMC11952393 DOI: 10.1101/2025.03.12.642696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Chronic inflammation and oral dysbiosis are common features of oral squamous cell carcinoma (OSCC). The commensal streptococci, S. anginosus, is increased in oral diseases including OSCC. Our previous work revealed that S. anginosus promotes inflammatory responses from macrophage cell lines, however the molecular mechanism by which S. anginosus interacts with macrophages to instigate this response remains to be investigated. Here, we expand on our previous findings by investigating the effects of S. anginosus infection of primary bone marrow derived macrophages (BMMs) and during in vivo infection. We found S. anginosus activated primary BMMs, which presented an enlarged cellular area, increased NF-κB activation and downstream inflammatory cytokines TNF⍰, IL-6 and IL-1β at 24 hours post infection. S. anginosus viability was dispensable for NF-κB activation, but essential for the induction of downstream inflammatory proteins and cytokines. S. anginosus persisted intracellularly within BMMs and induced the expression of inflammasome sensors AIM2, NLRC4 and NLRP3. Further, BMMs lacking the inflammasome adapter protein ASC ( Asc -/- ) had significantly diminished IL-1β production compared to wild type BMMs, indicating that S. anginosus activated the inflammasome. S. anginosus primarily triggered the inflammasome through NLRP3 as S. anginosus -infected Nlrp3 -/- BMMs and NLRP3 inhibitor (MCC950)-treated wild type BMMs displayed diminished IL-1β production compared to wild type controls. Lastly, S. anginosus -infected Asc -/- and Nlrp3 -/- mice displayed reduced weight loss compared to C57BL/6 mice. These overall findings indicate that S. anginosus replicates within macrophages and promotes a proinflammatory response in part through activation of the NLRP3 inflammasome. brief summary sentence: S. anginosus replicates intracellularly within macrophages and is sensed by the NLRP3 inflammasome to promote proinflammatory response.
Collapse
|
6
|
Lu J, Wu H, Wu S, Wang S, Fan H, Ruan H, Qiao J, Caiyin Q, Wen M. Salmonella: Infection mechanism and control strategies. Microbiol Res 2025; 292:128013. [PMID: 39675139 DOI: 10.1016/j.micres.2024.128013] [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: 08/22/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
Salmonella is a foodborne pathogen that predominantly resides in the intestinal tract of humans and animals. Infections caused by Salmonella can lead to various illnesses, including gastroenteritis, bacteremia, septicemia, and focal infections, with severe cases potentially resulting in host mortality. The mechanisms by which Salmonella invades host cells and disseminates throughout the body are partly understood, but there are still many scientific questions to be solved. This review aims to synthesize existing research on the interactions between Salmonella and hosts, detailing a comprehensive infection mechanism from adhesion and invasion to intracellular propagation and systemic spread. Overuse of antibiotics contributes to the emergence of drug-resistant Salmonella strains. An in-depth analysis of the mechanism of Salmonella infection will provide a theoretical basis for the development of novel Salmonella control strategies. These innovative control strategies include antibiotic adjuvants, small molecules, phages, attenuated vaccines, and probiotic therapies, which show huge potential in controlling Salmonella infection.
Collapse
Affiliation(s)
- Juane Lu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Hao Wu
- Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing 312300, China; School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Shengbo Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing 312300, China
| | - Shengli Wang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing 312300, China
| | - Hongfei Fan
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300072, China
| | - Haihua Ruan
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300072, China
| | - Jianjun Qiao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing 312300, China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Qinggele Caiyin
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China.
| | - Mingzhang Wen
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China; Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing 312300, China; Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300072, China.
| |
Collapse
|
7
|
Zhan L, Ge J, Xia L, Zhang Y. Reciprocal regulation between bacterial secretion systems and host metabolism: Enhancing bacterial intracellular survival capability. Microbiol Res 2025; 292:128025. [PMID: 39705830 DOI: 10.1016/j.micres.2024.128025] [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: 10/06/2024] [Revised: 11/23/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Secretion systems are intricate nanomachines present on many bacterial cell membranes that deliver various bacterially-encoded effector proteins into eukaryotic or prokaryotic cells. They are pivotal in bacterial invasion, host colonization, and pathogenesis. After infection, bacteria employ these machines to deliver toxic effectors to the cytoplasm of host cells that disrupt their metabolic balance, such as interfering with glucose metabolism, promoting lipid droplets formation, altering amino acid profiles and mitochondrial morphology, and reducing ROS levels, to ensure bacterial intracellular survival. Furthermore, metabolites within host cells can modulate the expression and/or function of bacterial secretion systems. This review summarizes recent advancements in understanding the impact of bacterial secretion systems on host cell metabolism and the feedback regulation of host metabolites on these machines, providing novel perspectives on host-pathogen interactions and mechanisms of bacterial pathogenesis.
Collapse
Affiliation(s)
- Lina Zhan
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, Zhenjiang, Jiangsu 212013, China
| | - Jiongchen Ge
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, Zhenjiang, Jiangsu 212013, China
| | - Lin Xia
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Ying Zhang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
8
|
Brancewicz J, Wójcik N, Sarnowska Z, Robak J, Król M. The Multifaceted Role of Macrophages in Biology and Diseases. Int J Mol Sci 2025; 26:2107. [PMID: 40076729 PMCID: PMC11900619 DOI: 10.3390/ijms26052107] [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: 02/05/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Macrophages are highly adaptable immune cells capable of responding dynamically to diverse environmental cues. They are pivotal in maintaining homeostasis, orchestrating immune responses, facilitating tissue repair, and, under certain conditions, contributing to disease pathogenesis. This review delves into the complex biology of macrophages, highlighting their polarization states, roles in autoimmune and inflammatory diseases, involvement in cancer progression, and potential as therapeutic targets. By understanding the context-dependent functional plasticity of macrophages, we can better appreciate their contributions to health and disease, paving the way for innovative therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | - Magdalena Król
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, Building 23, Level 0, Laboratory Number 0135, 8 Ciszewskiego St., 02-786 Warsaw, Poland
| |
Collapse
|
9
|
Emerson LE, Bhimani S, Rainey AL, Maurelli AT, Ferraro MJ. Evaluating small extracellular vesicle-based vaccination across heterologous Salmonella strains isolated from wastewater. Infect Immun 2025; 93:e0048524. [PMID: 39804074 PMCID: PMC11834434 DOI: 10.1128/iai.00485-24] [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: 11/05/2024] [Accepted: 12/04/2024] [Indexed: 02/13/2025] Open
Abstract
Salmonella infections pose significant public health challenges worldwide. The diversity of Salmonella strains, particularly those isolated from environmental and clinical sources, necessitates innovative approaches to prevention and treatment. Previous research has shown that small extracellular vesicles (sEVs) produced by macrophages during Salmonella Typhimurium infection can induce robust immune responses when used as a vaccine, offering complete protection in systemic infection models. In this study, we isolated 120 Salmonella strains from qPCR invA-positive wastewater samples collected in Gainesville, FL. These strains underwent enrichment, selection, and biochemical confirmation, followed by serotyping and whole genome sequencing. Two isolates, Salmonella enterica subsp. diarizonae (Diarizonae) and S. enterica serovar Enteritidis, were selected for further analysis based on community prevalence and clinical severity. We also assessed the ability of sEVs produced by S. Typhimurium-infected macrophages to induce immune responses against these heterologous and circulating strains in mice. Immunization with sEVs induced robust antigen-specific SIgA and IgG responses against S. Typhimurium, Enteritidis, and Diarizonae, with high titers observed in sera and fecal samples. Proteomic analysis revealed differential expression of proteins in these strains, including antigenic proteins present in sEVs such as OmpA, FliC, or OmpD. Moreover, this study highlights the role of wastewater-based epidemiology (WBE) as a tool for environmental surveillance, offering a complementary perspective on Salmonella dynamics within a population. Integrating WBE with traditional surveillance methods, along with the promising results of sEV-based vaccination, provides a pragmatic strategy for developing effective preventative measures against Salmonella infections, addressing the diversity of non-typhoidal Salmonella strains.
Collapse
Affiliation(s)
- Lisa E. Emerson
- Microbiology and Cell Science Department, IFAS, University of Florida, Gainesville, Florida, USA
- Clinical and Translational Science Institute, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Saloni Bhimani
- Microbiology and Cell Science Department, IFAS, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Andrew L. Rainey
- Clinical and Translational Science Institute, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Environmental and Global Health, University of Florida, Gainesville, Florida, USA
| | - Anthony T. Maurelli
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Environmental and Global Health, University of Florida, Gainesville, Florida, USA
| | - Mariola J. Ferraro
- Microbiology and Cell Science Department, IFAS, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
10
|
Yang Y, Zhou H, Li F, Zhang Y, Yang J, Shen Y, Hu N, Zou Q, Qin L, Zeng H, Huang W. Staphylococcus aureus induces mitophagy via the HDAC11/IL10 pathway to sustain intracellular survival. J Transl Med 2025; 23:156. [PMID: 39905391 DOI: 10.1186/s12967-025-06161-7] [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: 09/18/2024] [Accepted: 01/22/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND The immune evasion and prolonged survival of Staphylococcus aureus (S. aureus) within macrophages are key factors contributing to the difficulty in curing osteomyelitis. Although macrophages play a vital role as innate immune cells, the mechanisms by which S. aureus survives within them and suppresses host immune functions remain incompletely understood. METHODS This study employed confocal microscopy, flow cytometry, ELISA, and siRNA technology to assess the survival capacity of S. aureus within macrophages and the impact of inflammatory cytokines on its persistence. Proteomics was used to investigate the potential mechanisms and differential proteins involved in S. aureus intracellular survival. Additionally, confocal microscopy, flow cytometry, Mdivi-1 intervention, and Western blot were utilized to validate the role of mitophagy in supporting S. aureus survival. The study further explored how the HDAC11/IL10 axis enhances mitophagy to promote intracellular S. aureus survival by using HDAC11 overexpression, siRNA, and rapamycin intervention combined with confocal microscopy and flow cytometry. RESULTS The findings demonstrated that IL10 promotes mitophagy to clear mitochondrial reactive oxygen species (mtROS), thereby enhancing the intracellular survival of S. aureus within macrophages. Additionally, we discovered that the transcriptional repressor of IL10, HDAC11, was significantly downregulated during S. aureus infection. Overexpression of HDAC11 and the use of the autophagy activator rapamycin further validated that the HDAC11/IL10 axis regulates mitophagy via the mTOR pathway, which is essential for supporting S. aureus intracellular survival. CONCLUSION This study reveals that S. aureus enhances IL10 production by inhibiting HDAC11, thereby promoting mitophagy and mtROS clearance, which supports its survival within macrophages. These findings offer new insights into the intracellular survival mechanisms of S. aureus and provide potential therapeutic approaches for the clinical management of osteomyelitis.
Collapse
Affiliation(s)
- Yaji Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Haotian Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Feilong Li
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
- Department of Orthopaedics, The People's Hospital of Dazu, Chongqing, 402360, China
| | - Yanhao Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Jianye Yang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Yidong Shen
- Department of Orthopaedics, The First people's Hospital of Yancheng, Yancheng, Jiangsu, 224006, China
| | - Ning Hu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Leilei Qin
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China.
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China.
| | - Wei Huang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
11
|
Sweet MJ, Ramnath D, Singhal A, Kapetanovic R. Inducible antibacterial responses in macrophages. Nat Rev Immunol 2025; 25:92-107. [PMID: 39294278 DOI: 10.1038/s41577-024-01080-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 09/20/2024]
Abstract
Macrophages destroy bacteria and other microorganisms through phagocytosis-coupled antimicrobial responses, such as the generation of reactive oxygen species and the delivery of hydrolytic enzymes from lysosomes to the phagosome. However, many intracellular bacteria subvert these responses, escaping to other cellular compartments to survive and/or replicate. Such bacterial subversion strategies are countered by a range of additional direct antibacterial responses that are switched on by pattern-recognition receptors and/or host-derived cytokines and other factors, often through inducible gene expression and/or metabolic reprogramming. Our understanding of these inducible antibacterial defence strategies in macrophages is rapidly evolving. In this Review, we provide an overview of the broad repertoire of antibacterial responses that can be engaged in macrophages, including LC3-associated phagocytosis, metabolic reprogramming and antimicrobial metabolites, lipid droplets, guanylate-binding proteins, antimicrobial peptides, metal ion toxicity, nutrient depletion, autophagy and nitric oxide production. We also highlight key inducers, signalling pathways and transcription factors involved in driving these different antibacterial responses. Finally, we discuss how a detailed understanding of the molecular mechanisms of antibacterial responses in macrophages might be exploited for developing host-directed therapies to combat antibiotic-resistant bacterial infections.
Collapse
Affiliation(s)
- Matthew J Sweet
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.
| | - Divya Ramnath
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Amit Singhal
- Infectious Diseases Labs (ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Ronan Kapetanovic
- INRAE, Université de Tours, Infectiologie et Santé Publique (ISP), Nouzilly, France
| |
Collapse
|
12
|
Raj D, Nair AV, Singh A, Basu S, Sarkar K, Sharma J, Sharma S, Sharma S, Rathore M, Singh S, Prakash S, Simran, Sahu S, Kaushik AC, Siddiqi MI, Ghoshal UC, Chandra T, Bhosale V, Dasgupta A, Gupta SK, Verma S, Guha R, Chakravortty D, Ammanathan V, Lahiri A. Salmonella Typhimurium effector SseI regulates host peroxisomal dynamics to acquire lysosomal cholesterol. EMBO Rep 2025; 26:656-689. [PMID: 39695325 PMCID: PMC11811301 DOI: 10.1038/s44319-024-00328-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: 03/04/2024] [Revised: 10/16/2024] [Accepted: 10/25/2024] [Indexed: 12/20/2024] Open
Abstract
Salmonella enterica serotype Typhimurium (Salmonella) resides and multiplies intracellularly in cholesterol-rich compartments called Salmonella-containing vacuoles (SCVs) with actin-rich tubular extensions known as Salmonella-induced filaments (SIFs). SCV maturation depends on host-derived cholesterol, but the transport mechanism of low-density lipoprotein (LDL)-derived cholesterol to SCVs remains unclear. Here we find that peroxisomes are recruited to SCVs and function as pro-bacterial organelle. The Salmonella effector protein SseI is required for the interaction between peroxisomes and the SCV. SseI contains a variant of the PTS1 peroxisome-targeting sequence, GKM, localizes to the peroxisomes and activates the host Ras GTPase, ADP-ribosylation factor-1 (ARF-1). Activation of ARF-1 leads to the recruitment of phosphatidylinsolitol-5-phosphate-4 kinase and the generation of phosphatidylinsolitol-4-5-bisphosphate on peroxisomes. This enhances the interaction of peroxisomes with lysosomes and allows for the transfer of lysosomal cholesterol to SCVs using peroxisomes as a bridge. Salmonella infection of peroxisome-depleted cells leads to the depletion of cholesterol on the SCVs, resulting in reduced SIF formation and bacterial proliferation. Taken together, our work identified peroxisomes as a target of Salmonella secretory effectors, and as conveyance of host cholesterol to enhance SCV stability, SIF integrity, and intracellular bacterial growth.
Collapse
Affiliation(s)
- Desh Raj
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Abhilash Vijay Nair
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Anmol Singh
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Swarnali Basu
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Kabita Sarkar
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Jyotsna Sharma
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Shiva Sharma
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sanmi Sharma
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Manisha Rathore
- Laboratory Animal Facility Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shriya Singh
- Molecular Microbiology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shakti Prakash
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Simran
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Neuroscience & Ageing Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shikha Sahu
- Department of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medicine, Lucknow, India
| | - Aman Chandra Kaushik
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Mohammad Imran Siddiqi
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Biochemistry and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Uday C Ghoshal
- Department of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medicine, Lucknow, India
| | - Tulika Chandra
- Department of Transfusion Medicine, King Georges' Medical University, Lucknow, India
| | - Vivek Bhosale
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Toxicology and Experimental Medicine Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Arunava Dasgupta
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Molecular Microbiology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shashi Kumar Gupta
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sonia Verma
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Neuroscience & Ageing Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rajdeep Guha
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Laboratory Animal Facility Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India.
| | - Veena Ammanathan
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India.
| | - Amit Lahiri
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
13
|
Batsaikhan B, Lin PC, Shigemura K, Wu YW, Onishi R, Chang PR, Cheng HY, Fang SB. Comparison of global transcriptomes for nontyphoidal Salmonella clinical isolates from pediatric patients with and without bacteremia after their interaction with human intestinal epithelial cells in vitro. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2025; 58:38-47. [PMID: 39322508 DOI: 10.1016/j.jmii.2024.09.002] [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: 07/06/2023] [Revised: 08/30/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND Nontyphoidal Salmonella (NTS) outbreaks of invasive diseases are increasing. Whether the genetic diversity of invasive NTS correlates with the clinical characteristics and bacteremia development in NTS infections remains unclear. In this study, we compared the global transcriptomes between bacteremic and nonbacteremic NTS strains after their interaction with human intestinal epithelial cells in vitro. METHODS We selected clinical isolates obtained from stool and blood samples of patients with or without bacteremia and patients with high and low C-reactive protein (CRP) levels. The bacterial RNA samples were isolated after coculturing with Caco-2 cells for RNA sequencing and subsequent analyses. RESULTS CRP is an unreliable predictive maker for NTS bacteremia with a median CRP level of 1.6 mg/dL. Certain Salmonella Pathogenicity Island (SPI)-1 genes (sipC, sipA, sicA, sipD, and sipB), SPI-2 genes (ssaP, ssrA, and ssaS), and six SPI-4 genes (siiA, siiB, siiC, siiD, siiE, and siiF) remained upregulated in the bacteremic blood-derived strains but significantly downregulated in the nonbacteremic strains after their interaction with Caco-2 cells. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways analysis identified that arginine biosynthesis, ascorbate and aldarate metabolism, and phosphotransferase system pathways were activated in bacteremic NTS strains after Caco-2 cell priming. CONCLUSION CRP levels were not correlated with bacteremia development. Significant regulation of certain SPI genes in bacteremic NTS strains after Caco-2 cell priming; bacteremia development might be influenced by the host immune response and the extent to which specific metabolism pathways in NTS strains can be prevented from invading the bloodstream.
Collapse
Affiliation(s)
- Buyandelger Batsaikhan
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan
| | - Pei-Chun Lin
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Emergency Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Katsumi Shigemura
- Department of Public Health, Division of Infectious Diseases, Kobe University Graduate School of Health Sciences, Kobe, Japan; Department of Urology, Teikyo University School of Medicine, Tokyo, Japan
| | - Yu-Wei Wu
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, New Taipei City, Taiwan
| | - Reo Onishi
- Department of Public Health, Division of Infectious Diseases, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Pei-Ru Chang
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan
| | - Hung-Yen Cheng
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Shiuh-Bin Fang
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan; Master Program in Clinical Pharmacogenomics and Pharmacoproteomics, College of Pharmacy, Taipei Medical University, Taipei City, Taiwan; Research Center for Digestive Medicine, Taipei Medical University, Taipei City, Taiwan.
| |
Collapse
|
14
|
Zhou N, Ding Y, He T, Sun Y, Chen H, Huang M, Li T. Characterization and Protective Efficacy of a Salmonella Typhimurium ATCC 14028 sptP Mutant as a Live Attenuated Vaccine Candidate. Vaccines (Basel) 2025; 13:150. [PMID: 40006697 PMCID: PMC11860608 DOI: 10.3390/vaccines13020150] [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: 12/07/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Salmonella Typhimurium poses a substantial health risk to both humans and animals. This study evaluated the potential of using the Salmonella Typhimurium ΔsptP mutant as a live-attenuated vaccine candidate by constructing it through homologous recombination and assessing its key biological properties, including growth characteristics, immunogenicity, and protective efficacy. METHODS We generated the ΔsptP mutant through targeted gene deletion, ensuring the preservation of the bacterial strain's growth and stability. In vitro and in vivo assays were performed to compare the invasive capabilities between the mutant and the wild-type strains. Specifically, we examined the invasion into RAW264.7 murine macrophages and mice. Furthermore, the virulence of the mutant was evaluated by determining the median lethal dose (LD50). To evaluate immunogenicity and protection, mice were immunized with 2 × 104 CFUs of the ΔsptP mutant, followed by a booster immunization, and then challenged with a virulent strain. RESULTS The ΔsptP mutant exhibited no significant changes in growth characteristics or genetic stability compared to the wild-type strain. However, it demonstrated a significantly diminished capacity for invasion in both murine macrophages and mice. The LD50 for the mutant was 39.92-fold higher than that of the wild-type, indicating a marked reduction in virulence. Mice immunized with the ΔsptP mutant and administered a booster immunization exhibited 87.5% protection against challenge with a virulent strain, as compared to the PBS control group. Moreover, the mutant induced IgG antibody levels comparable to those induced by the wild-type strain. CONCLUSIONS The ΔsptP mutant of Salmonella Typhimurium exhibits markedly reduced virulence while retaining robust immunogenicity and protective efficacy. These findings suggest that the ΔsptP mutant is a promising candidate for a live-attenuated vaccine, potentially providing an effective strategy to prevent Salmonella Typhimurium infections.
Collapse
Affiliation(s)
- Nanlong Zhou
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (N.Z.)
| | - Yonghui Ding
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (N.Z.)
| | - Ting He
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (N.Z.)
| | - Yuling Sun
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (N.Z.)
| | - Hongfang Chen
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (N.Z.)
| | - Meiling Huang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou 571199, China
| | - Tiansen Li
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570100, China; (N.Z.)
| |
Collapse
|
15
|
Li M, Wang Y, Wang Q, Yang L, Liu S, Li G, Song Z, Huang C, Kang L, Zhang Y, Wang T, Kong L, Li S. A Mutant of Africa Swine Fever Virus Protein p72 Enhances Antibody Production and Regulates the Production of Cytokines. Viruses 2025; 17:194. [PMID: 40006949 PMCID: PMC11860850 DOI: 10.3390/v17020194] [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] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
African swine fever virus (ASFV) is a severe threat to the global pig industry, and domestic pigs mostly develop severe clinical manifestations upon viral invasion. Currently, there is no available vaccine against ASFV. Its capsid structural protein p72 is one of the immuno-dominant proteins. In this study, we unexpectedly obtained a p72 mutant protein (p72∆377-428) which deleted the aa 377-428 within p72 and had stable and high expression in E. coli. Using SWISS-MODEL 1.0 software, the prediction showed that p72∆377-428 was quite distinct from the wild-type p72 protein in structure. p72∆377-428 induced stronger antibody production in mice on day 42 and 56 post immunization and could recognize ASFV-infected swine sera. p72∆377-428 reduced IFN-γ production in the splenocytes from p72∆377-428-immunized mice and p72∆377-428-treated swine macrophages compared to p72. p72∆377-428 also decreased the production of pro-inflammatory cytokine genes, including IL-1β, IL-6, and IL-12, compared to p72 in mice. Further, we found that p72∆377-428 reduced the induction of pro-inflammatory cytokine genes by inhibiting AKT phosphorylation and HIF1α expression. Taken together, these findings have implications for immunological function and the corresponding mechanism of ASFV p72, and our study indicates that p72∆377-428 could serve as a novel candidate for ASFV vaccines and diagnostic reagents.
Collapse
Affiliation(s)
- Mingzhi Li
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330029, China; (M.L.)
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Nanchang 330029, China
- College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330029, China
| | - Yihao Wang
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330029, China; (M.L.)
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Nanchang 330029, China
- College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330029, China
| | - Quansheng Wang
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330029, China; (M.L.)
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Nanchang 330029, China
- College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330029, China
| | - Lingdi Yang
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330029, China; (M.L.)
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Nanchang 330029, China
- College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330029, China
| | - Shiguo Liu
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330029, China; (M.L.)
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Nanchang 330029, China
- College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330029, China
| | - Guangzhi Li
- College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330029, China
| | - Ziqi Song
- College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330029, China
| | - Chulu Huang
- College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330029, China
| | - Lumei Kang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330029, China
- Center for Laboratory Animal Science, Nanchang University, Nanchang 330031, China
| | - Yanni Zhang
- Jiangxi Province Center for Disease Control and Prevention, Nanchang 330029, China
| | - Ting Wang
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330029, China; (M.L.)
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Nanchang 330029, China
- College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330029, China
| | - Lingbao Kong
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330029, China; (M.L.)
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Nanchang 330029, China
- College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330029, China
| | - Sha Li
- Institute of Pathogenic Microorganism, Jiangxi Agricultural University, Nanchang 330029, China; (M.L.)
- Nanchang City Key Laboratory of Animal Virus and Genetic Engineering, Nanchang 330029, China
- College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang 330029, China
| |
Collapse
|
16
|
Santelices J, Schultz A, Walker A, Adams N, Tirado D, Barker H, Eshraghi A, Czyż DM, Ferraro MJ. Targeting deubiquitinating enzymes (DUBs) and ubiquitin pathway modulators to enhance host defense against bacterial infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635188. [PMID: 39975367 PMCID: PMC11838268 DOI: 10.1101/2025.01.27.635188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The rise of antibiotic-resistant bacterial pathogens poses a critical global health challenge, necessitating innovative therapeutic approaches. This study explores host-targeted therapies (HTTs) by focusing on deubiquitinating enzymes (DUBs), essential modulators of the ubiquitin-proteasome system (UPS) that regulate host-pathogen interactions during many bacterial infections. Using Salmonella-infected macrophages as a model, we identified UPS modulators that enhance bacterial clearance and observed significant changes in DUB expression, particularly USP25, USP46, and Otud7b. The small-molecule DUB inhibitor AZ-1 significantly reduced intracellular bacterial loads in vitro and mitigated early disease severity in a murine model by decreasing fecal bacterial loads and preserving host weight. However, AZ-1 alone did not achieve complete clearance of Salmonella and required combination with extracellular-targeting antibiotics for optimal efficacy. Notably, AZ-1 demonstrated broad-spectrum activity against multidrug-resistant pathogens, including Pseudomonas aeruginosa, Klebsiella pneumoniae, and Acinetobacter baumannii. Transcriptomic analyses revealed infection-induced DUB regulation and highlighted pathways modulating immune responses, including TNF-α secretion. These findings highlight the potential of targeting the UPS as a host-directed antimicrobial strategy and provide a foundation for developing innovative therapies to combat antimicrobial resistance.
Collapse
Affiliation(s)
- John Santelices
- Microbiology and Cell Science Department, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL 32611, USA
| | - Alexander Schultz
- Microbiology and Cell Science Department, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL 32611, USA
| | - Alyssa Walker
- Microbiology and Cell Science Department, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL 32611, USA
| | - Nicole Adams
- Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Deyaneira Tirado
- Microbiology and Cell Science Department, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL 32611, USA
| | - Hailey Barker
- Microbiology and Cell Science Department, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL 32611, USA
| | - Aria Eshraghi
- Department of Infectious Diseases & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Daniel M. Czyż
- Microbiology and Cell Science Department, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL 32611, USA
| | - Mariola J. Ferraro
- Microbiology and Cell Science Department, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
17
|
Ma Z, Ma J, Li J, Wang Z, Wei L, Ali A, Zuo Y, Cai X, Meng Q, Qiao J. Regulatory roles of the AraC family transcription factor yeaM in the virulence and biofilm formation of Salmonella Typhimurium. Int J Food Microbiol 2025; 431:111088. [PMID: 39893937 DOI: 10.1016/j.ijfoodmicro.2025.111088] [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: 10/15/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
Salmonella Typhimurium (S. typhimurium) is a significant zoonotic pathogen responsible for gastroenteritis and severe systemic infections in various hosts. The AraC family transcription factors are key gene expression regulators in prokaryotes, essential for bacterial adaptation to the environment and virulence. Despite their importance, the role of yeaM, a member of this family in S. typhimurium, remains unexplored. To elucidate yeaM regulatory function in virulence and biofilm formation, we engineered mutant and complementary strains of the yeaM gene using homologous recombination. We assessed their capabilities in biofilm formation under different conditions, macrophage adherence and invasion, and virulence in mice. Additionally, we identified potential target genes regulated by yeaM through transcriptome sequencing and confirmed these findings using an electrophoretic mobility shift assay (EMSA) and a dual-luciferase reporter assay. Our results demonstrate that, compared to the parental strain SL1344 and the complemented strain CΔyeaM, the ΔyeaM strain exhibited significantly enhanced biofilm formation, increased invasion of mouse intestinal epithelial cells, enhanced intracellular proliferation within macrophages, and elevated induction of macrophage apoptosis. Furthermore, the ΔyeaM deletion strain displayed significantly increased virulence in mice and enhanced proliferation in milk. Transcriptome analysis revealed that S. typhimurium pathogenicity island 4 (SPI4) genes (siiA, siiB, siiC, siiD, siiF, and siiE) were significantly upregulated following the deletion of the yeaM gene. EMSA and dual-luciferase reporter assays further showed that the yeaM protein can bind to the promoter of the siiA gene and suppress its expression, thereby modulating the biofilm formation and virulence of S. typhimurium.
Collapse
Affiliation(s)
- Zhongmei Ma
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Jifu Ma
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Jie Li
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Zhanpeng Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Lixiang Wei
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Ahmad Ali
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Yufei Zuo
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Xuepeng Cai
- State Key Lab of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu 730046, China
| | - Qingling Meng
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Jun Qiao
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| |
Collapse
|
18
|
Park J, Jang M, Choi E, Lee SM, Bang I, Woo J, Kim S, Lee EJ, Kim D. ChIP-mini: a low-input ChIP-exo protocol for elucidating DNA-binding protein dynamics in intracellular pathogens. Nucleic Acids Res 2025; 53:gkaf009. [PMID: 39868540 PMCID: PMC11770342 DOI: 10.1093/nar/gkaf009] [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: 07/25/2024] [Revised: 12/04/2024] [Accepted: 01/06/2025] [Indexed: 01/28/2025] Open
Abstract
Genome-wide identification of binding profiles for DNA-binding proteins from the limited number of intracellular pathogens in infection studies is crucial for understanding virulence and cellular processes but remains challenging, as the current ChIP-exo is designed for high-input bacterial cells (>1010). Here, we developed an optimized ChIP-mini method, a low-input ChIP-exo utilizing a 5,000-fold reduced number of initial bacterial cells and an analysis pipeline, to identify genome-wide binding dynamics of DNA-binding proteins in host-infected pathogens. Applying ChIP-mini to intracellular Salmonella Typhimurium, we identified 642 and 1,837 binding sites of H-NS and RpoD, respectively, elucidating changes in their binding position and binding intensity during infection. Post-infection, we observed 21 significant reductions in H-NS binding at intergenic regions, exposing the promoter region of virulence genes, such as those in Salmonella pathogenicity islands-2, 3 and effectors. Furthermore, we revealed the crucial phenomenon that novel and significantly increased RpoD bindings were found within regions exhibiting diminished H-NS binding, thereby facilitating substantial upregulation of virulence genes. These findings markedly enhance our understanding of how H-NS and RpoD simultaneously coordinate the transcription initiation of virulence genes within macrophages. Collectively, this work demonstrates a broadly adaptable tool that will enable the elucidation of DNA-binding protein dynamics in diverse intracellular pathogens during infection.
Collapse
Affiliation(s)
- Joon Young Park
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Minchang Jang
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Eunna Choi
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Sang-Mok Lee
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Ina Bang
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Jihoon Woo
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Seonggyu Kim
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Eun-Jin Lee
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Donghyuk Kim
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| |
Collapse
|
19
|
Dang Y, Sun X, Jiang J. [Role of Traditional Chinese Medicine in Pathogen-Induced Metabolic Reprogramming and Immune Suppression]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2025; 56:10-18. [PMID: 40109470 PMCID: PMC11914019 DOI: 10.12182/20250160501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Indexed: 03/22/2025]
Abstract
Traditional Chinese medicine (TCM), an essential component of the traditional medicine practiced in China, has demonstrated unique therapeutic efficacy in combating infectious diseases caused by pathogens and various types of tumors. In particular, TCM plays a vital role in enhancing immune function, maintaining homeostasis, and improving metabolic balance. However, the complex ingredients used in TCM and its broad range of therapeutic targets present challenges for comprehensive analysis of the mechanisms involved. Moreover, the molecular and cellular mechanisms underlying TCM's effects remain underexplored, limiting its broader application in modern medicine. Recent studies have increasingly revealed that TCM can not only directly inhibit the activity of pathogens, such as bacteria, fungi, viruses, and tumor cells, but also exert profound effects on immune remodeling by regulating the metabolism of both pathogens and hosts. Therefore, a comprehensive understanding of the role of TCM in metabolic regulation is crucial for elucidating its anti-pathogenic effects. This review is focused on the metabolic pathways of pathogens and host metabolic reprogramming induced by pathogens. We systematically reviewed the mechanisms by which TCM regulates pathogen metabolism, influences pathogen-induced metabolic reprogramming in hosts, and mitigates immune suppression caused by pathogens. This review may provide new ideas for investigating the molecular mechanisms of TCM in disease development and progression.
Collapse
Affiliation(s)
- Yixiong Dang
- / ( 610041) West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Xin Sun
- / ( 610041) West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
- ( 610041) Chinese Evidence-Based Medicine Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingwen Jiang
- / ( 610041) West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
20
|
Chen K, Wang J, Hu T, Zhao Y, Wu Y, Wang X, Li W, Yang G, Zhang L, Wang J, Zhu Y. Salmonella enterica serovar typhimurium effectors spiA and spiC promote replication by modulating iron metabolism and oxidative stress. Vet Microbiol 2025; 300:110328. [PMID: 39674031 DOI: 10.1016/j.vetmic.2024.110328] [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: 06/18/2024] [Revised: 10/22/2024] [Accepted: 11/30/2024] [Indexed: 12/16/2024]
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) poses a major threat to the health and safety of animal-derived foods worldwide. Recently, we have reported that S. Typhimurium uses iron to promote its own proliferation, leading to iron metabolism disorders. However, the mechanism by which S. Typhimurium induces iron metabolism disturbances remains unclear. In this study, we found that the S. Typhimurium effectors spiA and spiC promote the expression of iron regulatory protein 2 (IRP2), transferrin receptor 1 (TfR1) and divalent metal transporter protein 1 (DMT1) and inhibit the expression of ferroportin after transfection with the recombinant plasmids pEGFP-C1-spiA and pEGFP-C1-spiC, which in turn contributes to the accumulation of iron and oxidative stress. Furthermore, we aimed to verify the role of these two effector proteins in S. Typhimurium-induced disorders of iron metabolism. We constructed spiA or spiC mutant strains and their corresponding complementation strains. Our data showed that when spiA or spiC was knocked out, the upregulation of iron metabolism proteins (IRP2, TfR1 and DMT1), the accumulation of iron and oxidative stress caused by the wild-type strain were clearly alleviated in vitro and in vivo, which plays a key role in reducing the intracellular replication of S. Typhimurium and attenuating pathological damage to the liver and ileum of mice. Our findings highlighted that S. Typhimurium induces the disruption of iron metabolism via the virulence factors spiA and spiC, thereby facilitating S. Typhimurium proliferation and causing oxidative damage to the liver and ileum, which provides prospective insights into the search for effective antimicrobial targets for the defense against salmonellosis.
Collapse
Affiliation(s)
- Keyuan Chen
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Jing Wang
- Key Laboratory of Applied Biotechnology on Animal Science & Veterinary Medicine of Zhejiang Province, Zhejiang Engineering Research Center for Veterinary Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, Belt and Road International Joint Laboratory for One Health and Food Safety, China-Australia Joint Laboratory for Animal Health Big Data Analytics, College of Veterinary Medicine of Zhejiang A&F University, 666 Wusu Street, Lin'an District, Hangzhou, Zhejiang Province 311300, China.
| | - Ting Hu
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Yiqing Zhao
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Yi Wu
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Xue Wang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Wei Li
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Guiyan Yang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Linlin Zhang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Jiufeng Wang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| | - Yaohong Zhu
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China; College of Veterinary Medicine, Sanya Institute of China Agricultural University, Sanya 572000, China.
| |
Collapse
|
21
|
Ge Y, Zadeh M, Sharma C, Lin YD, Soshnev AA, Mohamadzadeh M. Controlling functional homeostasis of ileal resident macrophages by vitamin B12 during steady state and Salmonella infection in mice. Mucosal Immunol 2024; 17:1314-1325. [PMID: 39255854 DOI: 10.1016/j.mucimm.2024.08.011] [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: 06/25/2024] [Revised: 08/13/2024] [Accepted: 08/30/2024] [Indexed: 09/12/2024]
Abstract
Dietary micronutrients, particularly vitamin B12 (VB12), profoundly influence the physiological maintenance and function of intestinal cells. However, it is still unclear whether VB12 modulates the transcriptional and metabolic programming of ileal macrophages (iMacs), thereby contributing to intestinal homeostasis. Using multiomic approaches, we demonstrated that VB12 primarily supports the cell cycle activity and mitochondrial metabolism of iMacs, resulting in increased cell frequency compared to VB12 deficiency. VB12 also retained the ability to promote maintenance and metabolic regulation of iMacs during intestinal infection with Salmonella Typhimurium (STm). On the contrary, depletion of iMacs by inhibiting CSF1R signaling significantly increased host susceptibility to STm and prevented VB12-mediated pathogen reduction. These results thus suggest that regulation of VB12-dependent iMacs critically controls STm expansion, which may be of new relevance to advance our understanding of this vitamin and to strategically formulate sustainable therapeutic nutritional regimens that improve human gut health.
Collapse
Affiliation(s)
- Yong Ge
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health, San Antonio, TX, USA.
| | - Mojgan Zadeh
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health, San Antonio, TX, USA
| | - Cheshta Sharma
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health, San Antonio, TX, USA
| | - Yang-Ding Lin
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health, San Antonio, TX, USA
| | - Alexey A Soshnev
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas at San Antonio, TX, USA
| | - Mansour Mohamadzadeh
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health, San Antonio, TX, USA; South Texas Veterans Health Care System (STVHCS), San Antonio, TX, USA.
| |
Collapse
|
22
|
Wang X, Sun Y, Guo H, Yan X, Ma S, Yang B, Jiang L. Xylose utilization promotes Salmonella replication within macrophages and systemic infection in mice. Virulence 2024; 15:2435381. [PMID: 39603596 PMCID: PMC11610547 DOI: 10.1080/21505594.2024.2435381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/03/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024] Open
Abstract
The intracellular pathogen Salmonella can cause systemic diseases via its survival and replication in host macrophages. Xylose is the second most abundant sugar in nature and Salmonella can use xylose as its sole carbon source for growth. However, whether xylose utilization contributes to the pathogenicity and intracellular growth of Salmonella has not yet been determined. In this study, we observed that the xylose concentration in macrophages increased during Salmonella infection. Moreover, there was an increase in expression of Salmonella xylose catabolic genes (xylA and xylB) and the transcriptional regulatory gene of xylose metabolism (xylR) in macrophages, revealing the possibility of using host-accumulated xylose by Salmonella for intracellular growth. Mutation of either xylAB or xylR reduced Salmonella replication in macrophages and attenuated the colonization of mouse systemic loci (e.g. the liver and spleen), indicating that xylose utilization promotes Salmonella replication within macrophages and systemic infection in mice. Moreover, we found that xylose utilization by intracellular Salmonella was activated by the cAMP-CRP complex upon detection of low glucose levels in the infected macrophages. Collectively, these findings reveal that although the available glucose decreases during infection, Salmonella can use xylose, which accumulates in infected macrophages, as an alternative carbon source to promote intracellular replication and virulence.
Collapse
Affiliation(s)
- Xinyue Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Yuyang Sun
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Houliang Guo
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Xiaolin Yan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Shuai Ma
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Bin Yang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Lingyan Jiang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| |
Collapse
|
23
|
He H, Huang S, Geng N, Weng S, He J, Li C. Acute hypoxia stress mediates HIF-1α-Yki-Cactus axis to facilitate the infection of Vibrio parahaemolyticus in Litopenaeus vannamei. Front Immunol 2024; 15:1476309. [PMID: 39664389 PMCID: PMC11632965 DOI: 10.3389/fimmu.2024.1476309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
Introduction Hypoxia stress renders aquatic animals more susceptible to bacterial disease, yet the underlying mechanism remains elusive. Methods We conducted an acute hypoxia stress experiment to investigate the impact of stress on the immune response of Litopenaeus vannamei via transcriptome analysis, RT-qPCR and Western blot. Results Our results showed that acute hypoxia stress disrupted the tissue architecture, and significantly changed the gene expression profiles in the hepatopancreas of shrimp. More importantly, acute hypoxia stress significantly changed the expression levels of immune-related genes. Ladderlectin, GBP 1, Caspase-1, CLEC4F, MR1 and GBP 2 were significantly down-regulated, but HIF-1α, Cactus, TIPE, Akirin-2, Ivns1abp and TLR3 were significantly up-regulated. We further demonstrated that acute hypoxia activated Yki via HIF-1α to enhance expression level of Cactus, and then Cactus inhibited the phosphorylation of Dorsal and its nuclear translocation, thereby suppressing antibacterial immunity. Subsequently, the challenge experiment following stress revealed that exposure to acute hypoxia stress amplified the infectivity and lethality of Vibrio parahaemolyticus to shrimp. The mechanism of HIF-1α-Yki-Cautus axis provided an explanation for this phenomenon. Discussion This study offered new insights into interactions among environmental hypoxia stress, host immunity and pathogens, thereby providing practical guidelines for optimizing shrimp culture practices.
Collapse
Affiliation(s)
- Honghui He
- State Key Laboratory of Biocontrol/School of Marine Sciences, Sun Yat-sen University, Guangzhou, China
- Southern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, China
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangzhou, China
| | - Shaoqing Huang
- College of Marine Sciences, Beibu Gulf University, Qinzhou, China
| | - Ningze Geng
- State Key Laboratory of Biocontrol/School of Marine Sciences, Sun Yat-sen University, Guangzhou, China
- Southern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, China
| | - Shaoping Weng
- State Key Laboratory of Biocontrol/School of Marine Sciences, Sun Yat-sen University, Guangzhou, China
- Southern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, China
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangzhou, China
| | - Jianguo He
- State Key Laboratory of Biocontrol/School of Marine Sciences, Sun Yat-sen University, Guangzhou, China
- Southern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, China
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangzhou, China
| | - Chaozheng Li
- State Key Laboratory of Biocontrol/School of Marine Sciences, Sun Yat-sen University, Guangzhou, China
- Southern Marine Sciences and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, China
- School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangzhou, China
| |
Collapse
|
24
|
Yang L, Sun J, Yang T, Zhang X, Xu C, Wei Y, Li Y, Zhao Y, Zhang S, Wu Q, Shi H, Fu Q, Xia L. Therapeutic effects and mechanisms of berberine on enteritis caused by Salmonella in poultry. Front Microbiol 2024; 15:1458579. [PMID: 39664055 PMCID: PMC11631916 DOI: 10.3389/fmicb.2024.1458579] [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: 07/02/2024] [Accepted: 11/07/2024] [Indexed: 12/13/2024] Open
Abstract
The present study aimed to investigate the therapeutic effects of berberine (BBR) on Salmonella enteritis in broiler chickens and to elucidate its mechanisms of action preliminarily. Blood samples were collected from 21- to 35-day-old Sanhuang male chicks to measure immune and biochemical indicators and to calculate the organ coefficients for the liver, spleen, bursa of Fabricius, and thymus. The caecal microbiota was analysed through 16S ribosomal RNA (rRNA) gene sequencing, and transcriptome sequencing was conducted. Compared with the positive control group (S), the berberine-treated group (BS) presented increased serum immunoglobulin M (IgM) levels, serum IgG levels, and total antioxidant capacity; berberine ameliorated the increase in the thymus index caused by Salmonella administration. The addition of berberine to the diet increased the abundance of beneficial bacterial genera, including Bacteroides and Lactobacillus. It also decreased the abundance of harmful bacterial genera, including Faecalibacterium and Streptococcus. Transcriptome analysis revealed that gene expression in the S and BS groups was associated with T cell selection and B cell receptor signalling pathways, which are enriched primarily in multiple immune-related signalling pathways, including the B cell receptor signalling pathway, NF-κ B signalling pathway, intestinal immune network for IgA production, asthma, and African trypanosomiasis. The significantly expressed genes included ATAD5, ERP29, MGST2, PIK3CA, and HSP90AA1. The present study demonstrated that berberine has a good therapeutic effect on Salmonella infection in chicks, as it inhibits the occurrence and development of Salmonella-induced intestinal inflammation by regulating the balance of the gut microbiota and the expression of related genes, including ATAD5, ERP29, MGST2, PIK3CA, and HSP90AA1.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Lining Xia
- Xinjiang Key Laboratory of Herbivore Drug Research and Creation, College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang, China
| |
Collapse
|
25
|
Bao P, Zhang XZ. Progress of tumor-resident intracellular bacteria for cancer therapy. Adv Drug Deliv Rev 2024; 214:115458. [PMID: 39383997 DOI: 10.1016/j.addr.2024.115458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/12/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024]
Abstract
Emerging studies have disclosed the pivotal role of cancer-associated microbiota in supporting cancer development, progression and dissemination, with the in-depth comprehending of tumor microenvironment. In particular, certain invasive bacteria that hide in various cells within the tumor tissues can render assistance to tumor growth and invasion through intricate mechanisms implicated in multiple branches of cancer biology. Thus, tumor-resident intracellular microbes are anticipated as next-generation targets for oncotherapy. This review is intended to delve into these internalized bacteria-driven cancer-promoting mechanisms and explore diversified antimicrobial therapeutic strategies to counteract the detrimental impact caused by these intruders, thereby improving therapeutic benefit of antineoplastic therapy.
Collapse
Affiliation(s)
- Peng Bao
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital, Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, PR China
| | - Xian-Zheng Zhang
- Department of Orthopedic Trauma and Microsurgery of Zhongnan Hospital, Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, PR China.
| |
Collapse
|
26
|
Duan T, Li Z, Han X, Hong Q, Yang Y, Yan J, Xing C. Changes functional prediction of ear canal flora in chronic bacterial otitis externa. Front Cell Infect Microbiol 2024; 14:1434754. [PMID: 39507947 PMCID: PMC11538054 DOI: 10.3389/fcimb.2024.1434754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 09/24/2024] [Indexed: 11/08/2024] Open
Abstract
Objective To investigate ear canal microflora's structure, composition and function in patients with chronic bacterial otitis externa. Methods A case-control study design method was used to collect the ear canal secretions from 14 patients with chronic bacterial external otitis (CB group) and 14 healthy controls (H group) treated in the Department of Otolaryngology and Head and Neck Surgery in the First Affiliated Hospital of Hainan Medical University. 16S rRNA high-throughput sequencing technology was used to sequence the ear canal microflora's V3 ~ V4 region gene amplification products in the participating population. The α diversity of ear canal microflora in 2 groups was analyzed. Based on the weighted Unifrac distance, principal coordinate analysis was performed to compare the β diversity of ear tract microflora between the two groups. The differences in ear microflora at phylum and genus levels were analyzed. PICRUSt2 function prediction and BugBase phenotype prediction were also performed. Results α diversity analysis showed that the diversity and richness of auricular microflora in the CB group were significantly lower than those in the H group. β diversity analysis showed that there were some differences between the two groups. At the phylum level, the relative abundance of Bdellovibrionota, Campylobacterota, and WPS-2 in the microbiota of patients in the CB group was significantly lower than that in the H group, and the differences were statistically significant. At the genus level, the relative abundance of Pseudomonas, Acinetobacter, Pelomonas, Sphingomonas, Bradyrhizobium, Brevundimonas, Enhydrobacter, Actinomyces, Paracoccus and Chryseobacterium in the ear canal of Group H is significantly higher than that of Group CB. Functional prediction of PICRUSt2 suggests that amino acid biosynthesis and bacterial microbiota may be related. In BugBase phenotypic prediction, the contribution of aerobic phenotype in group CB was significantly lower than that in group H. Conclusion The diversity and abundance of the ear canal flora of patients with chronic bacterial otitis externa were significantly lower than those of the healthy population, and their bacterial colony structure was significantly altered. Dysbiosis of the ear canal flora may be an important cause of chronic bacterial otitis externa.
Collapse
Affiliation(s)
- Tingting Duan
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Zhiqun Li
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Xiaoyong Han
- Department of Otolaryngology, Linping District Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Qichao Hong
- Department of Otorhinolaryngology-Head and Neck Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Yunan Yang
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Jinren Yan
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| | - Chengliang Xing
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital, Hainan Medical University, Haikou, China
| |
Collapse
|
27
|
Graham CT, Gordon S, Kubes P. A historical perspective of Kupffer cells in the context of infection. Cell Tissue Res 2024:10.1007/s00441-024-03924-4. [PMID: 39392500 DOI: 10.1007/s00441-024-03924-4] [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: 06/28/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
The Kupffer cell was first discovered by Karl Wilhelm von Kupffer in 1876, labeling them as "Sternzellen." Since their discovery as the primary macrophages of the liver, researchers have gradually gained an in-depth understanding of the identity, functions, and influential role of Kupffer cells, particularly in infection. It is becoming clear that Kupffer cells perform important tissue-specific functions in homeostasis and disease. Stationary in the sinusoids of the liver, Kupffer cells have a high phagocytic capacity and are adept in clearing the bloodstream of foreign material, toxins, and pathogens. Thus, they are indispensable to host defense and prevent the dissemination of bacteria during infections. To highlight the importance of this cell, this review will explore the history of the Kupffer cell in the context of infection beginning with its discovery to the present day.
Collapse
Affiliation(s)
- Carolyn T Graham
- Department of Physiology and Pharmacology, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
| | - Siamon Gordon
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wenhua 1st Road Guishan Dist., Taoyuan, Taiwan
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| |
Collapse
|
28
|
Tang J, Wang X, Chen S, Chang T, Gu Y, Zhang F, Hou J, Luo Y, Li M, Huang J, Liu M, Zhang L, Wang Y, Shen X, Xu L. Disruption of glucose homeostasis by bacterial infection orchestrates host innate immunity through NAD +/NADH balance. Cell Rep 2024; 43:114648. [PMID: 39167491 DOI: 10.1016/j.celrep.2024.114648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/25/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
Metabolic reprogramming is crucial for activating innate immunity in macrophages, and the accumulation of immunometabolites is essential for effective defense against infection. The NAD+/NADH (ratio of nicotinamide adenine dinucleotide and its reduced counterpart) redox couple serves as a critical node that integrates metabolic pathways and signaling events, but how this metabolite couple engages macrophage activation remains unclear. Here, we show that the NAD+/NADH ratio serves as a molecular signal that regulates proinflammatory responses and type I interferon (IFN) responses divergently. Salmonella Typhimurium infection leads to a decreased NAD+/NADH ratio by inducing the accumulation of NADH. Further investigation shows that an increased NAD+/NADH ratio correlates with attenuated proinflammatory responses and enhanced type I IFN responses. Conversely, a decreased NAD+/NADH ratio is linked to intensified proinflammatory responses and restrained type I IFN responses. These results show that the NAD+/NADH ratio is an essential cell-intrinsic factor that orchestrates innate immunity, which enhances our understanding of how metabolites fine-tune innate immunity.
Collapse
Affiliation(s)
- Jingjing Tang
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiao Wang
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Shukun Chen
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Tianyuan Chang
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yanchao Gu
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Fuhua Zhang
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jing Hou
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yi Luo
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Mengyuan Li
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jianan Huang
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Mohua Liu
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Lei Zhang
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yao Wang
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xihui Shen
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Lei Xu
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
29
|
Liu X, Wang C, Gai W, Sun Z, Fang L, Hua Z. Critical role of msgA in invasive capacity and intracellular survivability of Salmonella. Appl Environ Microbiol 2024; 90:e0020124. [PMID: 39136487 PMCID: PMC11409701 DOI: 10.1128/aem.00201-24] [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: 02/05/2024] [Accepted: 07/04/2024] [Indexed: 09/19/2024] Open
Abstract
Salmonella enterica serovar Typhimurium, which is a common foodborne pathogen, causes both intestinal and systemic infections in hosts. Salmonella has a complex pathogenic mechanism that involves invasive capacity and intracellular survivability, which hampers research on virulence of Salmonella. The virulence of Salmonella is primarily studied through Salmonella pathogenicity islands (SPIs). However, there are also genes outside these SPIs that significantly impact virulence. Macrophage survival gene msgA is positioned at a region independent of the SPIs and conserved in Salmonella. However, there has been limited research on msgA to date. This study aims to investigate the virulent function of msgA to deepen our understanding of Salmonella virulence. Proteomic and RT-qPCR analyses reveal that MsgA influences multiple metabolic pathways and the expression of SPIs. The depletion of msgA led to the significantly reduced invasive capacity and intracellular survivability, and thus the decreased virulence of Salmonella. In conclusion, our study suggests that MsgA is an important regulator that mainly regulates virulence. Further research into the function of MsgA will enhance the understanding of Salmonella pathogenesis and promote the application of Salmonella for medical treatment. IMPORTANCE Salmonella enterica serovar Typhimurium is a common foodborne pathogen, it has a complex pathogenic mechanism that involves invasive capacity and intracellular survivability. The virulence of Salmonella is primarily studied through its pathogenicity islands. In contrast, virulence genes located outside the Salmonella pathogenicity islands (SPIs) have received less attention. Macrophage survival gene (MsgA) is positioned at a region independent of the SPIs and conserved in Salmonella. Our research indicates that MsgA is a novel global regulator influencing the metabolic pathways and SPIs. Further research into the function of MsgA will enhance the understanding of Salmonella pathogenesis and promote the application of Salmonella for medical treatment.
Collapse
Affiliation(s)
- Xinqi Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Chengzhi Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing, China
| | - Wenhua Gai
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Zhaotong Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Lei Fang
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing, China
| | - Zichun Hua
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc, Changzhou, China
| |
Collapse
|
30
|
Mayo-Pérez S, Gama-Martínez Y, Dávila S, Rivera N, Hernández-Lucas I. LysR-type transcriptional regulators: state of the art. Crit Rev Microbiol 2024; 50:598-630. [PMID: 37635411 DOI: 10.1080/1040841x.2023.2247477] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/29/2023]
Abstract
The LysR-type transcriptional regulators (LTTRs) are DNA-binding proteins present in bacteria, archaea, and in algae. Knowledge about their distribution, abundance, evolution, structural organization, transcriptional regulation, fundamental roles in free life, pathogenesis, and bacteria-plant interaction has been generated. This review focuses on these aspects and provides a current picture of LTTR biology.
Collapse
Affiliation(s)
- S Mayo-Pérez
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Y Gama-Martínez
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - S Dávila
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | - N Rivera
- IPN: CICATA, Unidad Morelos del Instituto Politécnico Nacional, Atlacholoaya, Mexico
| | - I Hernández-Lucas
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| |
Collapse
|
31
|
Saraiva MMS, Benevides VP, Guerra PR, Campos IC, Rodrigues Alves LB, Paiva JB, Muniz LM, Almeida AM, Freitas Neto OC, Olsen JE, Berchieri Junior A. Deletions of ttrA and pduA genes in Salmonella enterica affect survival within chicken-derived HD-11 macrophages. Curr Genet 2024; 70:14. [PMID: 39150461 DOI: 10.1007/s00294-024-01299-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/12/2024] [Accepted: 07/29/2024] [Indexed: 08/17/2024]
Abstract
In mammals, enteric salmonellas can use tetrathionate (ttr), formed as a by-product from the inflammatory process in the intestine, as electron acceptor in anaerobic respiration, and it can fuel its energy metabolism by degrading the microbial fermentation product 1,2-propanediol. However, recent studies have shown that this mechanism is not important for Salmonella infection in the intestine of poultry, while it prolongs the persistence of Salmonella at systemic sites in this species. In the current study, we show that ΔttrApduA strains of Salmonella enterica have lower net survival within chicken-derived HD-11 macrophages, as CFU was only 2.3% (S. Enteritidis ΔttrApduA), 2.3% (S. Heidelberg ΔttrApduA), and 3.0% (S. Typhimurium ΔttrApduA) compared to wild-type strains after 24 h inside HD-11 macrophage cells. The difference was not related to increased lysis of macrophages, and deletion of ttrA and pduA did not impair the ability of the strains to grow anaerobically. Further studies are indicated to determine the reason why Salmonella ΔttrApduA strains survive less well inside macrophage cell lines.
Collapse
Affiliation(s)
- Mauro M S Saraiva
- School of Agricultural and Veterinarian Sciences, São Paulo State University (UNESP), Jaboticabal, SP, 14884-900, Brazil.
- Department of Veterinary and Animal Sciences, University of Copenhagen (UK), Frederiksberg C., 1870, Denmark.
| | - Valdinete P Benevides
- School of Agricultural and Veterinarian Sciences, São Paulo State University (UNESP), Jaboticabal, SP, 14884-900, Brazil
- Department of Veterinary and Animal Sciences, University of Copenhagen (UK), Frederiksberg C., 1870, Denmark
| | - Priscila R Guerra
- Department of Veterinary and Animal Sciences, University of Copenhagen (UK), Frederiksberg C., 1870, Denmark
| | - Isabella C Campos
- School of Agricultural and Veterinarian Sciences, São Paulo State University (UNESP), Jaboticabal, SP, 14884-900, Brazil
| | - Lucas B Rodrigues Alves
- School of Agricultural and Veterinarian Sciences, São Paulo State University (UNESP), Jaboticabal, SP, 14884-900, Brazil
- Department of Veterinary and Animal Sciences, University of Copenhagen (UK), Frederiksberg C., 1870, Denmark
| | - Jacqueline B Paiva
- R&D Department, BioCamp Laboratories Ltda, Campinas, SP, 13082-020, Brazil
| | - Lauanda M Muniz
- R&D Department, BioCamp Laboratories Ltda, Campinas, SP, 13082-020, Brazil
| | - Adriana M Almeida
- School of Agricultural and Veterinarian Sciences, São Paulo State University (UNESP), Jaboticabal, SP, 14884-900, Brazil
| | - Oliveiro C Freitas Neto
- Department of Preventive Veterinary Medicine, Veterinary School, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, 31279-901, Brazil
| | - John E Olsen
- Department of Veterinary and Animal Sciences, University of Copenhagen (UK), Frederiksberg C., 1870, Denmark
| | - Angelo Berchieri Junior
- School of Agricultural and Veterinarian Sciences, São Paulo State University (UNESP), Jaboticabal, SP, 14884-900, Brazil.
| |
Collapse
|
32
|
Fei X, Li N, Xu X, Zhu Y. Macrophage biology in the pathogenesis of Helicobacter pylori infection. Crit Rev Microbiol 2024:1-18. [PMID: 39086061 DOI: 10.1080/1040841x.2024.2366944] [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: 08/03/2023] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 08/02/2024]
Abstract
Infection with H. pylori induces chronic gastric inflammation, progressing to peptic ulcer and stomach adenocarcinoma. Macrophages function as innate immune cells and play a vital role in host immune defense against bacterial infection. However, the distinctive mechanism by which H. pylori evades phagocytosis allows it to colonize the stomach and further aggravate gastric preneoplastic pathology. H. pylori exacerbates gastric inflammation by promoting oxidative stress, resisting macrophage phagocytosis, and inducing M1 macrophage polarization. M2 macrophages facilitate the proliferation, invasion, and migration of gastric cancer cells. Various molecular mechanisms governing macrophage function in the pathogenesis of H. pylori infection have been identified. In this review, we summarize recent findings of macrophage interactions with H. pylori infection, with an emphasis on the regulatory mechanisms that determine the clinical outcome of bacterial infection.
Collapse
Affiliation(s)
- Xiao Fei
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Nianshuang Li
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xinbo Xu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yin Zhu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
33
|
Zhang Z, Wang Y, Xia L, Zhang Y. Roles of Critical Amino Acids Metabolism in The Interactions Between Intracellular Bacterial Infection and Macrophage Function. Curr Microbiol 2024; 81:280. [PMID: 39031203 DOI: 10.1007/s00284-024-03801-x] [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: 05/03/2024] [Accepted: 07/10/2024] [Indexed: 07/22/2024]
Abstract
Macrophages, as crucial participants in the innate immune system, respond to pathogenic challenges through their dynamic metabolic adjustments, demonstrating the intimate interplay between cellular metabolism and immune function. Bacterial infection of macrophages causes changes in macrophage metabolism, affecting both macrophage function and bacterial virulence and intracellular survival. This review explores the reprogramming of amino acid metabolism in macrophages in response to bacterial infection, with a particular focus on the influence of critical amino acids such as serine, glutamine, and arginine on the immune functions of macrophages; highlights the roles of these metabolic pathways in macrophage functions such as phagocytosis, inflammatory response, immune regulation, and pathogen clearance; reveals how pathogens exploit and manipulate the amino acid metabolism within macrophages to support their own growth and replication, thereby showcasing the intricate interplay between macrophages and pathogens. It provides a foundation for understanding the interactions between macrophages amino acid metabolism and pathogens, offering potential strategies and therapeutic targets for the development of novel anti-infection therapies.
Collapse
Affiliation(s)
- Zuowei Zhang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, Zhenjiang, 212013, Jiangsu, China
- International Genome Center, Jiangsu University, Zhenjiang, 212013, China
| | - Yurou Wang
- International Genome Center, Jiangsu University, Zhenjiang, 212013, China
| | - Lin Xia
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Ying Zhang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
34
|
Jin H, Chen Y, Zhang D, Lin J, Huang S, Wu X, Deng W, Huang J, Yao Y. YTHDF2 favors protumoral macrophage polarization and implies poor survival outcomes in triple negative breast cancer. iScience 2024; 27:109902. [PMID: 38812540 PMCID: PMC11134561 DOI: 10.1016/j.isci.2024.109902] [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: 12/24/2023] [Revised: 03/11/2024] [Accepted: 05/01/2024] [Indexed: 05/31/2024] Open
Abstract
Patients with triple-negative breast cancer (TNBC) frequently experience resistance to chemotherapy, leading to recurrence. The approach of optimizing anti-tumoral immunological effect is promising in overcoming such resistance, given the heterogeneity and lack of biomarkers in TNBC. In this study, we focused on YTHDF2, an N6-methyladenosine (m6A) RNA-reader protein, in macrophages, one of the most abundant intra-tumoral immune cells. Using single-cell sequencing and ex vivo experiments, we discovered that YTHDF2 significantly promotes pro-tumoral phenotype polarization of macrophages and is closely associated with down-regulated antigen-presentation signaling to other immune cells in TNBC. The in vitro deprivation of YTHDF2 favors anti-tumoral effect. Expressions of multiple transcription factors, especially SPI1, were consistently observed in YTHDF2-high macrophages, providing potential therapeutic targets for new strategies. In conclusion, YTHDF2 in macrophages appears to promote pro-tumoral effects while suppressing immune activity, indicating the treatment targeting YTHDF2 or its transcription factors could be a promising strategy for chemoresistant TNBC.
Collapse
Affiliation(s)
- Hao Jin
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
| | - Yue Chen
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
| | - Dongbo Zhang
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
| | - Junfan Lin
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
| | - Songyin Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
| | - Xiaohua Wu
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
| | - Wen Deng
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
| | - Jiandong Huang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong Province 518055, China
- Clinical Oncology Center, Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Yandan Yao
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510120, China
- Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province 516621, China
- Guangdong Provincial Key Laboratory of Cancer Pathogenesis and Precision Diagnosis and Treatment, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province 516621, China
| |
Collapse
|
35
|
Parada-Kusz M, Clatworthy AE, Goering ER, Blackwood SM, Shigeta JY, Mashin E, Salm EJ, Choi C, Combs S, Lee JSW, Rodriguez-Osorio C, Clish C, Tomita S, Hung DT. 3-Hydroxykynurenine targets kainate receptors to promote defense against infection. Nat Chem Biol 2024:10.1038/s41589-024-01635-z. [PMID: 38898166 DOI: 10.1038/s41589-024-01635-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 05/07/2024] [Indexed: 06/21/2024]
Abstract
Bacterial infection involves a complex interaction between the pathogen and host where the outcome of infection is not solely determined by pathogen eradication. To identify small molecules that promote host survival by altering the host-pathogen dynamic, we conducted an in vivo chemical screen using zebrafish embryos and found that treatment with 3-hydroxykynurenine (3-HK) protects from lethal bacterial infection. 3-HK, a metabolite produced through host tryptophan metabolism, has no direct antibacterial activity but enhances host survival by restricting bacterial expansion in macrophages through a systemic mechanism that targets kainate-sensitive glutamate receptors. These findings reveal a new pathway by which tryptophan metabolism and kainate-sensitive glutamate receptors function and interact to modulate immunity, with important implications for the coordination between the immune and nervous systems in pathological conditions.
Collapse
Affiliation(s)
- Margarita Parada-Kusz
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anne E Clatworthy
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA.
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Emily R Goering
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Stephanie M Blackwood
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jack Y Shigeta
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Elizabeth J Salm
- Department of Cellular and Molecular Physiology and Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Catherine Choi
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Senya Combs
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jenny S W Lee
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carlos Rodriguez-Osorio
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Clary Clish
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Susumu Tomita
- Department of Cellular and Molecular Physiology and Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Deborah T Hung
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital (MGH), Boston, MA, USA.
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
36
|
Cottam C, White RT, Beck LC, Stewart CJ, Beatson SA, Lowe EC, Grinter R, Connolly JPR. Metabolism of L-arabinose converges with virulence regulation to promote enteric pathogen fitness. Nat Commun 2024; 15:4462. [PMID: 38796512 PMCID: PMC11127945 DOI: 10.1038/s41467-024-48933-7] [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: 12/18/2023] [Accepted: 05/16/2024] [Indexed: 05/28/2024] Open
Abstract
Virulence and metabolism are often interlinked to control the expression of essential colonisation factors in response to host-associated signals. Here, we identified an uncharacterised transporter of the dietary monosaccharide ʟ-arabinose that is widely encoded by the zoonotic pathogen enterohaemorrhagic Escherichia coli (EHEC), required for full competitive fitness in the mouse gut and highly expressed during human infection. Discovery of this transporter suggested that EHEC strains have an enhanced ability to scavenge ʟ-arabinose and therefore prompted us to investigate the impact of this nutrient on pathogenesis. Accordingly, we discovered that ʟ-arabinose enhances expression of the EHEC type 3 secretion system, increasing its ability to colonise host cells, and that the underlying mechanism is dependent on products of its catabolism rather than the sensing of ʟ-arabinose as a signal. Furthermore, using the murine pathogen Citrobacter rodentium, we show that ʟ-arabinose metabolism provides a fitness benefit during infection via virulence factor regulation, as opposed to supporting pathogen growth. Finally, we show that this mechanism is not restricted to ʟ-arabinose and extends to other pentose sugars with a similar metabolic fate. This work highlights the importance integrating central metabolism with virulence regulation in order to maximise competitive fitness of enteric pathogens within the host-niche.
Collapse
Affiliation(s)
- Curtis Cottam
- Newcastle University Biosciences Institute, Newcastle University, NE2 4HH, Newcastle-upon-Tyne, UK
| | - Rhys T White
- Institute of Environmental Science and Research, Wellington, New Zealand
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Lauren C Beck
- Newcastle University Translation and Clinical Research Institute, Newcastle University, NE2 4HH, Newcastle-upon-Tyne, UK
| | - Christopher J Stewart
- Newcastle University Translation and Clinical Research Institute, Newcastle University, NE2 4HH, Newcastle-upon-Tyne, UK
| | - Scott A Beatson
- Australian Infectious Diseases Research Centre and School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Elisabeth C Lowe
- Newcastle University Biosciences Institute, Newcastle University, NE2 4HH, Newcastle-upon-Tyne, UK
| | - Rhys Grinter
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - James P R Connolly
- Newcastle University Biosciences Institute, Newcastle University, NE2 4HH, Newcastle-upon-Tyne, UK.
| |
Collapse
|
37
|
Wódz K, Piechowicz L, Tokarska-Pietrzak E, Gawor J, Gromadka R, Bełkot Z, Strzałkowska Z, Wiśniewski J, Nowak T, Bogdan J, Anusz K, Pławińska-Czarnak J. Does Salmonella diarizonae 58:r:z 53 Isolated from a Mallard Duck Pose a Threat to Human Health? Int J Mol Sci 2024; 25:5664. [PMID: 38891852 PMCID: PMC11171591 DOI: 10.3390/ijms25115664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 06/21/2024] Open
Abstract
Salmonella diarizonae (IIIb) is frequently isolated from reptiles and less frequently from birds and mammals. However, its isolation from invasive human infections has not been widely reported. Migratory mallard ducks are excellent bioindicators of pathogen presence and pathogen antibiotic resistance (AMR). We present the first isolation from a mallard duck in central Europe of the antibiotic-resistant Salmonella enterica subsp. diarizonae with the unique antigenic pattern 58:r:z53 and report its whole-genome sequencing, serosequencing, and genotyping, which enabled the prediction of its pathogenicity and comparison with phenotypic AMR. The isolated strain was highly similar to S. diarizonae isolated from humans and food. Twenty-four AMR genes were detected, including those encoding aminoglycoside, fluoroquinolone, macrolide, carbapenem, tetracycline, cephalosporin, nitroimidazole, peptide antibiotic, and disinfecting agent/antiseptic resistance. Six Salmonella pathogenicity islands were found (SPI-1, SPI-2, SPI-3, SPI-5, SPI-9, and SPI-13). An iron transport system was detected in SPI-1 centisome C63PI. Plasmid profile analyses showed three to be present. Sequence mutations in the invA and invF genes were noted, which truncated and elongated the proteins, respectively. The strain also harbored genes encoding type-III secretion-system effector proteins and many virulence factors found in S. diarizonae associated with human infections. This study aims to elucidate the AMR and virulence genes in S. enterica subsp. diarizonae that may most seriously threaten human health.
Collapse
Affiliation(s)
- Karolina Wódz
- Laboratory of Molecular Biology, Vet-Lab Brudzew, 62-720 Brudzew, Poland;
| | - Lidia Piechowicz
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdańsk, 80-204 Gdańsk, Poland; (L.P.); (E.T.-P.)
| | - Ewa Tokarska-Pietrzak
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdańsk, 80-204 Gdańsk, Poland; (L.P.); (E.T.-P.)
| | - Jan Gawor
- DNA Sequencing and Synthesis Facility, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland; (J.G.); (R.G.)
| | - Robert Gromadka
- DNA Sequencing and Synthesis Facility, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland; (J.G.); (R.G.)
| | - Zbigniew Bełkot
- Department of Food Hygiene of Animal Origin, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, 20-950 Lublin, Poland;
| | - Zuzanna Strzałkowska
- Department of Food Hygiene and Public Health Protection, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland; (Z.S.); (J.W.); (J.B.); (K.A.)
| | - Jan Wiśniewski
- Department of Food Hygiene and Public Health Protection, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland; (Z.S.); (J.W.); (J.B.); (K.A.)
| | - Tomasz Nowak
- Laboratory of Molecular Biology, Vet-Lab Brudzew, 62-720 Brudzew, Poland;
| | - Janusz Bogdan
- Department of Food Hygiene and Public Health Protection, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland; (Z.S.); (J.W.); (J.B.); (K.A.)
| | - Krzysztof Anusz
- Department of Food Hygiene and Public Health Protection, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland; (Z.S.); (J.W.); (J.B.); (K.A.)
| | - Joanna Pławińska-Czarnak
- Department of Food Hygiene and Public Health Protection, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland; (Z.S.); (J.W.); (J.B.); (K.A.)
| |
Collapse
|
38
|
Machado LFM, Galán JE. Loss of function of metabolic traits in typhoidal Salmonella without apparent genome degradation. mBio 2024; 15:e0060724. [PMID: 38572992 PMCID: PMC11077982 DOI: 10.1128/mbio.00607-24] [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: 02/28/2024] [Accepted: 03/12/2024] [Indexed: 04/05/2024] Open
Abstract
Salmonella enterica serovar Typhi and Paratyphi A are the cause of typhoid and paratyphoid fever in humans, which are systemic life-threatening illnesses. Both serovars are exclusively adapted to the human host, where they can cause life-long persistent infection. A distinct feature of these serovars is the presence of a relatively high number of degraded coding sequences coding for metabolic pathways, most likely a consequence of their adaptation to a single host. As a result of convergent evolution, these serovars shared many of the degraded coding sequences although often affecting different genes in the same metabolic pathway. However, there are several coding sequences that appear intact in one serovar while clearly degraded in the other, suggesting differences in their metabolic capabilities. Here, we examined the functionality of metabolic pathways that appear intact in S. Typhi but that show clear signs of degradation in S. Paratyphi A. We found that, in all cases, the existence of single amino acid substitutions in S. Typhi metabolic enzymes, transporters, or transcription regulators resulted in the inactivation of these metabolic pathways. Thus, the inability of S. Typhi to metabolize Glucose-6-Phosphate or 3-phosphoglyceric acid is due to the silencing of the expression of the genes encoding the transporters for these compounds due to point mutations in the transcriptional regulatory proteins. In contrast, its inability to utilize glucarate or galactarate is due to the presence of point mutations in the transporter and enzymes necessary for the metabolism of these sugars. These studies provide additional support for the concept of adaptive convergent evolution of these two human-adapted S. enterica serovars and highlight a limitation of bioinformatic approaches to predict metabolic capabilities. IMPORTANCE Salmonella enterica serovar Typhi and Paratyphi A are the cause of typhoid and paratyphoid fever in humans, which are systemic life-threatening illnesses. Both serovars can only infect the human host, where they can cause life-long persistent infection. Because of their adaptation to the human host, these bacterial pathogens have changed their metabolism, leading to the loss of their ability to utilize certain nutrients. In this study we examined the functionality of metabolic pathways that appear intact in S. Typhi but that show clear signs of degradation in S. Paratyphi A. We found that, in all cases, the existence of single amino acid substitutions in S. Typhi metabolic enzymes, transporters, or transcription regulators resulted in the inactivation of these metabolic pathways. These studies provide additional support for the concept of adaptive convergent evolution of these two human-adapted S. enterica serovars.
Collapse
Affiliation(s)
- Leopoldo F. M. Machado
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jorge E. Galán
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
39
|
Achi SC, McGrosso D, Tocci S, Ibeawuchi SR, Sayed IM, Gonzalez DJ, Das S. Proteome profiling identifies a link between the mitochondrial pathways and host-microbial sensor ELMO1 following Salmonella infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592405. [PMID: 38746404 PMCID: PMC11092768 DOI: 10.1101/2024.05.03.592405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The host EnguLfment and cell MOtility protein 1 (ELMO1) is a cytosolic microbial sensor that facilitates bacterial sensing, internalization, clearance, and inflammatory responses. We have shown previously that ELMO1 binds bacterial effector proteins, including pathogenic effectors from Salmonella and controls host innate immune signaling. To understand the ELMO1-regulated host pathways, we have performed liquid chromatography Multinotch MS3-Tandem Mass Tag (TMT) multiplexed proteomics to determine the global quantification of proteins regulated by ELMO1 in macrophages during Salmonella infection. Comparative proteome analysis of control and ELMO1-depleted murine J774 macrophages after Salmonella infection quantified more than 7000 proteins with a notable enrichment in mitochondrial-related proteins. Gene ontology enrichment analysis revealed 19 upregulated and 11 downregulated proteins exclusive to ELMO1-depleted cells during infection, belonging to mitochondrial functions, metabolism, vesicle transport, and the immune system. By assessing the cellular energetics via Seahorse analysis, we found that Salmonella infection alters mitochondrial metabolism, shifting it from oxidative phosphorylation to glycolysis. Importantly, these metabolic changes are significantly influenced by the depletion of ELMO1. Furthermore, ELMO1 depletion resulted in a decreased ATP rate index following Salmonella infection, indicating its importance in counteracting the effects of Salmonella on immunometabolism. Among the proteins involved in mitochondrial pathways, mitochondrial fission protein DRP1 was significantly upregulated in ELMO1-depleted cells and in ELMO1-KO mice intestine following Salmonella infection. Pharmacological Inhibition of DRP1 revealed the link of the ELMO1-DRP1 pathway in regulating the pro-inflammatory cytokine TNF-α following infection. The role of ELMO1 has been further characterized by a proteome profile of ELMO1-depleted macrophage infected with SifA mutant and showed the involvement of ELMO1-SifA on mitochondrial function, metabolism and host immune/defense responses. Collectively, these findings unveil a novel role for ELMO1 in modulating mitochondrial functions, potentially pivotal in modulating inflammatory responses. Significance Statement Host microbial sensing is critical in infection and inflammation. Among these sensors, ELMO1 has emerged as a key regulator, finely tuning innate immune signaling and discriminating between pathogenic and non-pathogenic bacteria through interactions with microbial effectors like SifA of Salmonella . In this study, we employed Multinotch MS3-Tandem Mass Tag (TMT) multiplexed proteomics to determine the proteome alterations mediated by ELMO1 in macrophages following WT and SifA mutant Salmonella infection. Our findings highlight a substantial enrichment of host proteins associated with metabolic pathways and mitochondrial functions. Notably, we validated the mitochondrial fission protein DRP1 that is upregulated in ELMO1-depleted macrophages and in ELMO1 knockout mice intestine after infection. Furthermore, we demonstrated that Salmonella -induced changes in cellular energetics are influenced by the presence of ELMO1. This work shed light on a possible novel link between mitochondrial dynamics and microbial sensing in modulating immune responses.
Collapse
|
40
|
Su M, Yin M, Zhou Y, Xiao S, Yi J, Tang R. Freeze-Thaw Microfluidic System Produces "Themis" Nanocomplex for Cleaning Persisters-Infected Macrophages and Enhancing Uninfected Macrophages. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311436. [PMID: 38181783 DOI: 10.1002/adma.202311436] [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: 10/31/2023] [Revised: 01/01/2024] [Indexed: 01/07/2024]
Abstract
Macrophages are the primary effectors against potential pathogen infections. They can be "parasitized" by intracellular bacteria, serving as "accomplices", protecting intracellular bacteria and even switching them to persisters. Here, using a freeze-thaw strategy-based microfluidic chip, a "Themis" nanocomplex (TNC) is created. The TNC consists of Lactobacillus reuteri-derived membrane vesicles, heme, and vancomycin, which cleaned infected macrophages and enhanced uninfected macrophages. In infected macrophages, TNC releases heme that led to the reconstruction of the respiratory chain complexes of intracellular persisters, forcing them to regrow. The revived bacteria produces virulence factors that destroyed host macrophages (accomplices), thereby being externalized and becoming vulnerable to immune responses. In uninfected macrophages, TNC upregulates the TCA cycle and oxidative phosphorylation (OXPHOS), contributing to immunoenhancement. The combined effect of TNC of cleaning the accomplice (infected macrophages) and reinforcing uninfected macrophages provides a promising strategy for intracellular bacterial therapy.
Collapse
Affiliation(s)
- Mingyue Su
- School of stomatology, Lanzhou University, Lanzhou, 730000, China
| | - Mengying Yin
- School of stomatology, Lanzhou University, Lanzhou, 730000, China
| | - Yifu Zhou
- School of stomatology, Lanzhou University, Lanzhou, 730000, China
| | - Shuya Xiao
- School of stomatology, Lanzhou University, Lanzhou, 730000, China
| | - Jundan Yi
- School of stomatology, Lanzhou University, Lanzhou, 730000, China
| | - Rongbing Tang
- School of stomatology, Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
41
|
Du W, Li S, Li F. Different Immune Responses of Hemocytes from V. parahaemolyticus-Resistant and -Susceptible Shrimp at Early Infection Stage. BIOLOGY 2024; 13:300. [PMID: 38785782 PMCID: PMC11117703 DOI: 10.3390/biology13050300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 04/16/2024] [Accepted: 04/21/2024] [Indexed: 05/25/2024]
Abstract
Vibrio parahaemolyticus is one of the main causative agents leading to acute hepatopancreatic necrosis disease, the severe bacterial disease that occurs during shrimp aquaculture. Hemocytes play important roles during Vibrio infection. Previously, we found that there were few differentially expressed genes (DEGs) between hemocytes from V. parahaemolyticus-resistant and -susceptible shrimp before infection. We considered that there should be different immune responses between them after a pathogen infection. Here, the transcriptome data of hemocytes from V. parahaemolyticus-resistant and -susceptible shrimp before and after a pathogen infection were compared. The results showed that there were 157 DEGs responsive to infection in V. parahaemolyticus-resistant shrimp, while 33 DEGs in V. parahaemolyticus-susceptible shrimp. DEGs in V. parahaemolyticus-resistant shrimp were mainly related to immune and glycolytic processes, while those in V. parahaemolyticus-susceptible shrimp were mainly related to metabolism, with only two DEGs in common. A further analysis of genes involved in glucose metabolism revealed that GLUT2, HK, FBP, and PCK1 were lowly expressed while PC were highly expressed in hemocytes of the V. parahaemolyticus-resistant shrimp, indicating that glucose metabolism in shrimp hemocytes was related to a V. parahaemolyticus infection. After the knockdown of PC, the expression of genes in Toll and IMD signaling pathways were down-regulated, indicating that glucose metabolism might function through regulating host immunity during V. parahaemolyticus infection. The results suggest that the immune responses between V. parahaemolyticus-resistant and -susceptible shrimp were apparently different, which probably contribute to their different V. parahaemolyticus resistance abilities.
Collapse
Affiliation(s)
- Wenran Du
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao 266109, China;
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China;
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Shihao Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China;
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan 430072, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
| | - Fuhua Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China;
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan 430072, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
| |
Collapse
|
42
|
Junaid M, Lu H, Din AU, Yu B, Liu Y, Li Y, Liu K, Yan J, Qi Z. Deciphering Microbiome, Transcriptome, and Metabolic Interactions in the Presence of Probiotic Lactobacillus acidophilus against Salmonella Typhimurium in a Murine Model. Antibiotics (Basel) 2024; 13:352. [PMID: 38667028 PMCID: PMC11047355 DOI: 10.3390/antibiotics13040352] [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: 02/19/2024] [Revised: 04/06/2024] [Accepted: 04/08/2024] [Indexed: 04/29/2024] Open
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium), a foodborne pathogen that poses significant public health risks to humans and animals, presents a formidable challenge due to its antibiotic resistance. This study explores the potential of Lactobacillus acidophilus (L. acidophilus 1.3251) probiotics as an alternative strategy to combat antibiotic resistance associated with S. Typhimurium infection. In this investigation, twenty-four BALB/c mice were assigned to four groups: a non-infected, non-treated group (CNG); an infected, non-treated group (CPG); a group fed with L. acidophilus but not infected (LAG); and a group fed with L. acidophilus and challenged with Salmonella (LAST). The results revealed a reduction in Salmonella levels in the feces of mice, along with restored weight and improved overall health in the LAST compared to the CPG. The feeding of L. acidophilus was found to downregulate pro-inflammatory cytokine mRNA induced by Salmonella while upregulating anti-inflammatory cytokines. Additionally, it influenced the expression of mRNA transcript, encoding tight junction protein, oxidative stress-induced enzymes, and apoptosis-related mRNA expression. Furthermore, the LEfSe analysis demonstrated a significant shift in the abundance of critical commensal genera in the LAST, essential for maintaining gut homeostasis, metabolic reactions, anti-inflammatory responses, and butyrate production. Transcriptomic analysis revealed 2173 upregulated and 506 downregulated differentially expressed genes (DEGs) in the LAST vs. the CPG. Functional analysis of these DEGs highlighted their involvement in immunity, metabolism, and cellular development. Kyoto Encyclopedia of Genes and Genome (KEGG) pathway analysis indicated their role in tumor necrosis factor (TNF), mitogen-activated protein kinase (MAPK), chemokine, Forkhead box O (FOXO), and transforming growth factor (TGF-β) signaling pathway. Moreover, the fecal metabolomic analysis identified 929 differential metabolites, with enrichment observed in valine, leucine, isoleucine, taurine, glycine, and other metabolites. These findings suggest that supplementation with L. acidophilus promotes the growth of beneficial commensal genera while mitigating Salmonella-induced intestinal disruption by modulating immunity, gut homeostasis, gut barrier integrity, and metabolism.
Collapse
Affiliation(s)
| | - Hongyu Lu
- Medical College, Guangxi University, Nanning 530004, China
| | - Ahmad Ud Din
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC 28081, USA
| | - Bin Yu
- Medical College, Guangxi University, Nanning 530004, China
| | - Yu Liu
- Medical College, Guangxi University, Nanning 530004, China
| | - Yixiang Li
- Medical College, Guangxi University, Nanning 530004, China
| | - Kefei Liu
- Tianjin Shengji Group., Co., Ltd., No. 2, Hai Tai Development 2nd Road, Huayuan Industrial Zone, Tianjin 300384, China
| | - Jianhua Yan
- Medical College, Guangxi University, Nanning 530004, China
| | - Zhongquan Qi
- Medical College, Guangxi University, Nanning 530004, China
| |
Collapse
|
43
|
Gu D, Li A, Zang X, Huang T, Guo Y, Jiao X, Pan Z. Salmonella Enteritidis antitoxin DinJ inhibits NLRP3-dependent canonical inflammasome activation in macrophages. Infect Immun 2024; 92:e0050523. [PMID: 38477589 PMCID: PMC11003228 DOI: 10.1128/iai.00505-23] [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: 12/06/2023] [Accepted: 02/09/2024] [Indexed: 03/14/2024] Open
Abstract
The inflammasome is a pivotal component of the innate immune system, acting as a multiprotein complex that plays an essential role in detecting and responding to microbial infections. Salmonella Enteritidis have evolved multiple mechanisms to regulate inflammasome activation and evade host immune system clearance. Through screening S. Enteritidis C50336ΔfliC transposon mutant library, we found that the insertion mutant of dinJ increased inflammasome activation. In this study, we demonstrated the genetic connection between the antitoxin DinJ and the toxin YafQ in S. Enteritidis, confirming their co-transcription. The deletion mutant ΔfliCΔdinJ increased cell death and IL-1β secretion in J774A.1 cells. Western blotting analysis further showed elevated cleaved Caspase-1 product (p10 subunits) and IL-1β secretion in cells infected with ΔfliCΔdinJ compared to cells infected with ΔfliC. DinJ was found to inhibit canonical inflammasome activation using primary bone marrow-derived macrophages (BMDMs) from Casp-/- C57BL/6 mice. Furthermore, DinJ specifically inhibited NLRP3 inflammasome activation, as demonstrated in BMDMs from Nlrp3-/- and Nlrc4-/- mice. Fluorescence resonance energy transfer (FRET) experiments confirmed the translocation of DinJ into host cells during infection. Finally, we revealed that DinJ could inhibit the secretion of IL-1β and IL-18 in vivo, contributing to S. Enteritidis evading host immune clearance. In summary, our findings provide insights into the role of DinJ in modulating the inflammasome response during S. Enteritidis infection, highlighting its impact on inhibiting inflammasome activation and immune evasion.
Collapse
Affiliation(s)
- Dan Gu
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ang Li
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xirui Zang
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu, China
| | - Tingting Huang
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yaxin Guo
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhiming Pan
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, China
- Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
44
|
Kumar M, Sharma S, Kumar J, Barik S, Mazumder S. Mitochondrial electron transport chain in macrophage reprogramming: Potential role in antibacterial immune response. CURRENT RESEARCH IN IMMUNOLOGY 2024; 5:100077. [PMID: 38572399 PMCID: PMC10987323 DOI: 10.1016/j.crimmu.2024.100077] [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: 11/17/2023] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024] Open
Abstract
Macrophages restrain microbial infection and reinstate tissue homeostasis. The mitochondria govern macrophage metabolism and serve as pivot in innate immunity, thus acting as immunometabolic regulon. Metabolic pathways produce electron flows that end up in mitochondrial electron transport chain (mtETC), made of super-complexes regulating multitude of molecular and biochemical processes. Cell-intrinsic and extrinsic factors influence mtETC structure and function, impacting several aspects of macrophage immunity. These factors provide the macrophages with alternate fuel sources and metabolites, critical to gain functional competence and overcoming pathogenic stress. Mitochondrial reactive oxygen species (mtROS) and oxidative phosphorylation (OXPHOS) generated through the mtETC are important innate immune attributes, which help macrophages in mounting antibacterial responses. Recent studies have demonstrated the role of mtETC in governing mitochondrial dynamics and macrophage polarization (M1/M2). M1 macrophages are important for containing bacterial pathogens and M2 macrophages promote tissue repair and wound healing. Thus, mitochondrial bioenergetics and metabolism are intimately coupled with innate immunity. In this review, we have addressed mtETC function as innate rheostats that regulate macrophage reprogramming and innate immune responses. Advancement in this field encourages further exploration and provides potential novel macrophage-based therapeutic targets to control unsolicited inflammation.
Collapse
Affiliation(s)
- Manmohan Kumar
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Shagun Sharma
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
- Department of Zoology, Gargi College, University of Delhi, Delhi, India
| | - Jai Kumar
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Sailen Barik
- EonBio, 3780 Pelham Drive, Mobile, AL 36619, USA
| | - Shibnath Mazumder
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
- Faculty of Life Sciences and Biotechnology, South Asian University, Delhi, India
| |
Collapse
|
45
|
Almeida L, Dhillon-LaBrooy A, Sparwasser T. The evolutionary tug-of-war of macrophage metabolism during bacterial infection. Trends Endocrinol Metab 2024; 35:235-248. [PMID: 38040578 DOI: 10.1016/j.tem.2023.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 12/03/2023]
Abstract
The function and phenotype of macrophages are intimately linked with pathogen detection. On sensing pathogen-derived signals and molecules, macrophages undergo a carefully orchestrated process of polarization to acquire pathogen-clearing properties. This phenotypic change must be adequately supported by metabolic reprogramming that is now known to support the acquisition of effector function, but also generates secondary metabolites with direct microbicidal activity. At the same time, bacteria themselves have adapted to both manipulate and take advantage of macrophage-specific metabolic adaptations. Here, we summarize the current knowledge on macrophage metabolism during infection, with a particular focus on understanding the 'arms race' between host immune cells and bacteria during immune responses.
Collapse
Affiliation(s)
- Luís Almeida
- Institute of Medical Microbiology and Hygiene, University Medical Center of Johannes Gutenberg University, Mainz 55131, Germany.
| | - Ayesha Dhillon-LaBrooy
- Institute of Medical Microbiology and Hygiene, University Medical Center of Johannes Gutenberg University, Mainz 55131, Germany
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene, University Medical Center of Johannes Gutenberg University, Mainz 55131, Germany.
| |
Collapse
|
46
|
Wenner N, Zhu X, Rowe WPM, Händler K, Hinton JCD. Succinate utilisation by Salmonella is inhibited by multiple regulatory systems. PLoS Genet 2024; 20:e1011142. [PMID: 38457455 DOI: 10.1371/journal.pgen.1011142] [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: 01/17/2024] [Revised: 03/26/2024] [Accepted: 01/19/2024] [Indexed: 03/10/2024] Open
Abstract
Succinate is a potent immune signalling molecule that is present in the mammalian gut and within macrophages. Both of these infection niches are colonised by the pathogenic bacterium Salmonella enterica serovar Typhimurium during infection. Succinate is a C4-dicarboyxlate that can serve as a source of carbon for bacteria. When succinate is provided as the sole carbon source for in vitro cultivation, Salmonella and other enteric bacteria exhibit a slow growth rate and a long lag phase. This growth inhibition phenomenon was known to involve the sigma factor RpoS, but the genetic basis of the repression of bacterial succinate utilisation was poorly understood. Here, we use an experimental evolution approach to isolate fast-growing mutants during growth of S. Typhimurium on succinate containing minimal medium. Our approach reveals novel RpoS-independent systems that inhibit succinate utilisation. The CspC RNA binding protein restricts succinate utilisation, an inhibition that is antagonised by high levels of the small regulatory RNA (sRNA) OxyS. We discovered that the Fe-S cluster regulatory protein IscR inhibits succinate utilisation by repressing the C4-dicarboyxlate transporter DctA. Furthermore, the ribose operon repressor RbsR is required for the complete RpoS-driven repression of succinate utilisation, suggesting a novel mechanism of RpoS regulation. Our discoveries shed light on the redundant regulatory systems that tightly regulate the utilisation of succinate. We speculate that the control of central carbon metabolism by multiple regulatory systems in Salmonella governs the infection niche-specific utilisation of succinate.
Collapse
Affiliation(s)
- Nicolas Wenner
- Clinical Infection, Microbiology & Immunology, Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Xiaojun Zhu
- Clinical Infection, Microbiology & Immunology, Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Will P M Rowe
- Clinical Infection, Microbiology & Immunology, Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Kristian Händler
- Department of Microbiology, School of Genetics and Microbiology, Moyne Institute of Preventive Medicine, Trinity College Dublin, Dublin, Ireland
| | - Jay C D Hinton
- Clinical Infection, Microbiology & Immunology, Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
47
|
Chen YT, Lohia GK, Chen S, Riquelme SA. Immunometabolic Regulation of Bacterial Infection, Biofilms, and Antibiotic Susceptibility. J Innate Immun 2024; 16:143-158. [PMID: 38310854 PMCID: PMC10914382 DOI: 10.1159/000536649] [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: 07/26/2023] [Accepted: 02/01/2024] [Indexed: 02/06/2024] Open
Abstract
BACKGROUND Upon infection, mucosal tissues activate a brisk inflammatory response to clear the pathogen, i.e., resistance to disease. Resistance to disease is orchestrated by tissue-resident macrophages, which undergo profound metabolic reprogramming after sensing the pathogen. These metabolically activated macrophages release many inflammatory factors, which promote their bactericidal function. However, in immunocompetent individuals, pathogens like Pseudomonas aeruginosa, Staphylococcus aureus, and Salmonella evade this type of immunity, generating communities that thrive for the long term. SUMMARY These organisms develop features that render them less susceptible to eradication, such as biofilms and increased tolerance to antibiotics. Furthermore, after antibiotic therapy withdrawal, "persister" cells rapidly upsurge, triggering inflammatory relapses that worsen host health. How these pathogens persisted in inflamed tissues replete with activated macrophages remains poorly understood. KEY MESSAGES In this review, we discuss recent findings indicating that the ability of P. aeruginosa, S. aureus, and Salmonella to evolve biofilms and antibiotic tolerance is promoted by the similar metabolic routes that regulate macrophage metabolic reprogramming.
Collapse
Affiliation(s)
- Ying-Tsun Chen
- Department of Pediatrics, Division of Infectious Diseases, Columbia University, New York, New York, USA
| | - Gaurav Kumar Lohia
- Department of Pediatrics, Division of Infectious Diseases, Columbia University, New York, New York, USA
| | - Samantha Chen
- Department of Pediatrics, Division of Infectious Diseases, Columbia University, New York, New York, USA
| | - Sebastián A Riquelme
- Department of Pediatrics, Division of Infectious Diseases, Columbia University, New York, New York, USA
| |
Collapse
|
48
|
Augenstreich J, Shuster M, Fan Y, Lyu Z, Ling J, Briken V. BBQ methods: streamlined workflows for bacterial burden quantification in infected cells by confocal microscopy. Biol Open 2024; 13:bio060189. [PMID: 38156988 PMCID: PMC10836645 DOI: 10.1242/bio.060189] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024] Open
Abstract
Accurate quantification of bacterial burden within macrophages, termed bacterial burden quantification (BBQ), is crucial for understanding host-pathogen interactions. Various methods have been employed, each with strengths and weaknesses. This article addresses limitations in existing techniques and introduces two novel, automated methods for BBQ within macrophages based on confocal microscopy data analysis. The first method refines total fluorescence quantification by incorporating filtering steps to exclude uninfected cells, while the second method calculates total bacterial volume per cell to mitigate potential biases in fluorescence-based readouts. These workflows utilize PyImageJ and Cellpose software, providing reliable, unbiased, and rapid quantification of bacterial load. The proposed workflows were validated using Salmonella enterica serovar Typhimurium and Mycobacterium tuberculosis models, demonstrating their effectiveness in accurately assessing bacterial burden. These automated workflows offer valuable tools for studying bacterial interactions within host cells and provide insights for various research applications.
Collapse
Affiliation(s)
- Jacques Augenstreich
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Michael Shuster
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Yongqiang Fan
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Zhihui Lyu
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Jiqiang Ling
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Volker Briken
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
49
|
Wu L, Du Z, Li L, Qiao L, Zhang S, Yin X, Chang X, Li C, Hua Z. Camouflaging attenuated Salmonella by cryo-shocked macrophages for tumor-targeted therapy. Signal Transduct Target Ther 2024; 9:14. [PMID: 38195682 PMCID: PMC10776584 DOI: 10.1038/s41392-023-01703-1] [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: 09/08/2023] [Revised: 10/17/2023] [Accepted: 11/08/2023] [Indexed: 01/11/2024] Open
Abstract
Live bacteria-mediated antitumor therapies mark a pivotal point in cancer immunotherapy. However, the difficulty in reconciling the safety and efficacy of bacterial therapies has limited their application. Improving bacterial tumor-targeted delivery while maintaining biosafety is a critical hurdle for the clinical translation of live microbial therapy for cancer. Here, we developed "dead" yet "functional" Salmonella-loaded macrophages using liquid nitrogen cold shock of an attenuated Salmonella typhimurium VNP20009-contained macrophage cell line. The obtained "dead" macrophages achieve an average loading of approximately 257 live bacteria per 100 cells. The engineered cells maintain an intact cellular structure but lose their original pathogenicity, while intracellular bacteria retain their original biological activity and are delay freed, followed by proliferation. This "Trojan horse"-like bacterial camouflage strategy avoids bacterial immunogenicity-induced neutrophil recruitment and activation in peripheral blood, reduces the clearance of bacteria by neutrophils and enhances bacterial tumor enrichment efficiently after systemic administration. Furthermore, this strategy also strongly activated the tumor microenvironment, including increasing antitumor effector cells (including M1-like macrophages and CD8+ Teffs) and decreasing protumor effector cells (including M2-like macrophages and CD4+ Tregs), and ultimately improved antitumor efficacy in a subcutaneous H22 tumor-bearing mouse model. The cryo-shocked macrophage-mediated bacterial delivery strategy holds promise for expanding the therapeutic applications of living bacteria for cancer.
Collapse
Affiliation(s)
- Leyang Wu
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Neurology of Nanjing Drum Tower Hospital, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, 21008, Jiangsu, China
- Nanjing Generecom Biotechnology Co., Ltd, Nanjing, 210023, China
- Changzhou High-Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc, Changzhou, 213164, Jiangsu, China
| | - Zengzheng Du
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Neurology of Nanjing Drum Tower Hospital, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, 21008, Jiangsu, China
| | - Lin Li
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Neurology of Nanjing Drum Tower Hospital, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, 21008, Jiangsu, China
| | - Liyuan Qiao
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Neurology of Nanjing Drum Tower Hospital, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, 21008, Jiangsu, China
| | - Shuhui Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Neurology of Nanjing Drum Tower Hospital, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, 21008, Jiangsu, China
| | - Xingpeng Yin
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Neurology of Nanjing Drum Tower Hospital, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, 21008, Jiangsu, China
| | - Xiaoyao Chang
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Neurology of Nanjing Drum Tower Hospital, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, 21008, Jiangsu, China
| | - Chenyang Li
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Neurology of Nanjing Drum Tower Hospital, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, 21008, Jiangsu, China
| | - Zichun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology and Department of Neurology of Nanjing Drum Tower Hospital, School of Life Sciences and The Affiliated Hospital of Nanjing University Medical School, Nanjing University, Nanjing, 21008, Jiangsu, China.
- Nanjing Generecom Biotechnology Co., Ltd, Nanjing, 210023, China.
- Changzhou High-Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc, Changzhou, 213164, Jiangsu, China.
- School of Biopharmacy, China Pharmaceutical University, Nanjing, 210023, Jiangsu, China.
| |
Collapse
|
50
|
Wang X, Yang B, Ma S, Yan X, Ma S, Sun H, Sun Y, Jiang L. Lactate promotes Salmonella intracellular replication and systemic infection via driving macrophage M2 polarization. Microbiol Spectr 2023; 11:e0225323. [PMID: 37796020 PMCID: PMC10715217 DOI: 10.1128/spectrum.02253-23] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/21/2023] [Indexed: 10/06/2023] Open
Abstract
IMPORTANCE The important enteropathogen Salmonella can cause lethal systemic infection via survival and replication in host macrophages. Lactate represents an abundant intracellular metabolite during bacterial infection, which can also induce macrophage M2 polarization. In this study, we found that macrophage-derived lactate promotes the intracellular replication and systemic infection of Salmonella. During Salmonella infection, lactate via the Salmonella type III secretion system effector SteE promotes macrophage M2 polarization, and the induction of macrophage M2 polarization by lactate is responsible for lactate-mediated Salmonella growth promotion. This study highlights the complex interactions between Salmonella and macrophages and provides an additional perspective on host-pathogen crosstalk at the metabolic interface.
Collapse
Affiliation(s)
- Xinyue Wang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Bin Yang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Shuangshuang Ma
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
- Department of Biopharmaceuticals, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xiaolin Yan
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Shuai Ma
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Hongmin Sun
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Yuyang Sun
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Lingyan Jiang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| |
Collapse
|