1
|
Du LY, Keerthisinghe P, Rolland L, Sung YJ, Darroch H, Linnerz T, Ashimbayeva E, Grant MJ, Kakadia PM, Ramachandran A, Tups A, Spaink HP, Bohlander SK, Cheeseman J, Crosier PS, Astin JW, Warman G, Hall CJ. A light-regulated circadian timer optimizes neutrophil bactericidal activity to boost daytime immunity. Sci Immunol 2025; 10:eadn3080. [PMID: 40408429 DOI: 10.1126/sciimmunol.adn3080] [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: 12/03/2023] [Revised: 01/21/2025] [Accepted: 04/30/2025] [Indexed: 05/25/2025]
Abstract
The immune response exhibits strong circadian rhythmicity, with enhanced bacterial clearance often synchronized with an organism's active phase. Despite providing the bulk of cellular antibacterial defense, the neutrophil clockwork is poorly understood. Here, we used larval zebrafish to explore the role of clock genes in neutrophils during infection. Per2 was required in neutrophils for reactive oxygen species (ROS) production and bacterial killing by enhancing infection-responsive expression of high-mobility group box 1a (hmgb1a). The Cry binding domain of Per2 was required for regulation of neutrophil bactericidal activity, and neutrophils lacking Cry1a had elevated bactericidal activity and infection-responsive hmgb1a expression. A conserved cis-regulatory element with BMAL1 and nuclear factor κB binding motifs gated infection-responsive hmgb1a expression to the light phase. Mutagenesis of the BMAL1 motif in neutrophils blunted the priming effect of light on bactericidal activity and hmgb1a expression. These findings identify a light-responsive cell-intrinsic timer that controls time-of-day variations in antibacterial activity.
Collapse
Affiliation(s)
- Lucia Yi Du
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Biozentrum, University of Basel, Basel, Switzerland
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | - Pramuk Keerthisinghe
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Centre For Neuroendocrinology, University of Otago, Dunedin, New Zealand
| | - Leah Rolland
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Yih Jian Sung
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Hannah Darroch
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Tanja Linnerz
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Elina Ashimbayeva
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | | | - Purvi M Kakadia
- Leukaemia & Blood Cancer Research Unit, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Annasuya Ramachandran
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Alexander Tups
- Centre For Neuroendocrinology, University of Otago, Dunedin, New Zealand
| | - Herman P Spaink
- Institute of Biology, Leiden University, Leiden, Netherlands
| | - Stefan K Bohlander
- Leukaemia & Blood Cancer Research Unit, Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - James Cheeseman
- Department of Anaesthesiology, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Philip S Crosier
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Jonathan W Astin
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Guy Warman
- Department of Anaesthesiology, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Christopher J Hall
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
2
|
Cheng HY, Chu J, Limjunyawong N, Chen J, Ye Y, Chen KH, Koylass N, Sun S, Dong X, Qiu Z. Phagosome-mediated anti-bacterial immunity is governed by the proton-activated chloride channel in peritoneal macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640612. [PMID: 40060571 PMCID: PMC11888413 DOI: 10.1101/2025.02.27.640612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
The success of phagosome degradation relies on the ability of phagocytes to regulate the maturation of phagosomes. However, its underlying molecular mechanisms remain poorly understood. Here, we identify the proton-activated chloride (PAC) channel as a key negative regulator of phagosome maturation. PAC deletion enhanced phagosomal acidification and protease activities, leading to augmented bacterial killing in large peritoneal macrophages (LPMs) upon Escherichia coli infection in mice. Surprisingly, phagosome degradation also stimulated STING-IRF3-interferon responses and inflammasome activation in LPMs, both of which are enhanced upon PAC deletion. The increased inflammasome activation induced the release of cleaved gasdermin D, which localized to the surface of bacteria in the peritoneum and further contributed to their killing. Finally, enhanced bacterial clearance by PAC-deficient LPMs reduced proinflammatory immune cell infiltration and peritoneal inflammation, resulting in improved survival in mice. Our study thus provides new insights into the molecular mechanism of phagosome maturation and the dynamics of host defense response following phagosome-mediated bacterial degradation in peritoneal macrophages. Summary The PAC channel mediates phagosome maturation during bacterial infection in macrophages. PAC deletion promotes phagosome-mediated STING-interferon signaling and inflammasome-mediated gasdermin D secretion during bacterial infection in peritoneal macrophages.
Collapse
|
3
|
Cagnoli CI, Chiapparrone ML, Acuña F, Cacciato CS, Rodríguez MG, Aller JF, Catena MDC. Characterization of Campylobacter fetus adherence, invasiveness, and ultrastructural damage on bovine oviductal cells. Microb Pathog 2025; 202:107429. [PMID: 40021032 DOI: 10.1016/j.micpath.2025.107429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Bovine campylobacteriosis is a distributed worldwide disease caused by Campylobacter fetus. It is a sexual transmitted disease that affects reproductive health in cattle. The objective of this study was to use bovine oviduct primary cell cultures as a pathogenicity model to study the virulence of Campylobacter fetus fetus and Campylobacter fetus venerealis. Both subspecies showed the ability to adhere to, invade and cause cytopathogenic effect in oviductal cells. Transmission electron microscopy revealed significant ultrastructural damage, including cytoplasmic vacuolization, nuclear condensation and mitochondrial alterations. These mitochondrial changes, such enlargement and fragmentation, suggest potential impacts on cell viability and host cellular metabolism. This study is the first to investigate the pathogenicity of both subspecies in primary bovine oviductal epithelial cells. These findings demonstrate the virulence mechanism of C. fetus in the reproductive tract, providing insights into oviductal pathogenicity and potential impacts on bovine fertility.
Collapse
Affiliation(s)
- Claudia Inés Cagnoli
- Laboratorio de Microbiología Clínica y Experimental, Centro de Investigación Veterinaria de Tandil (CIVETAN) (UNCPBA-CICPBA-CONICET), Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, Tandil, Buenos Aires, Argentina.
| | - María Laura Chiapparrone
- Laboratorio de Microbiología Clínica y Experimental, Centro de Investigación Veterinaria de Tandil (CIVETAN) (UNCPBA-CICPBA-CONICET), Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, Tandil, Buenos Aires, Argentina.
| | - Francisco Acuña
- Laboratorio de Histología y Embriología Descriptiva, Experimental y Comparada, Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, Buenos Aires, Argentina.
| | - Claudio Santiago Cacciato
- Laboratorio de Microbiología Clínica y Experimental, Centro de Investigación Veterinaria de Tandil (CIVETAN) (UNCPBA-CICPBA-CONICET), Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, Tandil, Buenos Aires, Argentina.
| | - Marcelo Gastón Rodríguez
- Área de Bioestadística, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, Tandil, Buenos Aires, Argentina.
| | - Juan Florencio Aller
- Biotecnología de la Reproducción, Departamento de Producción Animal, Estación Experimental Agropecuaria Balcarce, Instituto Nacional de Tecnología Agropecuaria (INTA), Balcarce, Buenos Aires, Argentina.
| | - María Del Carmen Catena
- Laboratorio de Microbiología Clínica y Experimental, Centro de Investigación Veterinaria de Tandil (CIVETAN) (UNCPBA-CICPBA-CONICET), Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, Tandil, Buenos Aires, Argentina.
| |
Collapse
|
4
|
Herta T, Bhattacharyya A, Hippenstiel S, Zahlten J. The role of KLF4 in phagocyte activation during infectious diseases. Front Immunol 2025; 16:1584873. [PMID: 40313940 PMCID: PMC12044337 DOI: 10.3389/fimmu.2025.1584873] [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: 02/27/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
Phagocytes, including granulocytes (especially neutrophils), monocytes, macrophages, and dendritic cells, are essential components of the innate immune system, bridging innate and adaptive immunity. Their activation and function are tightly regulated by transcription factors that coordinate immune responses. Among these, Krüppel-like factor 4 (KLF4) has gained attention as a regulator of phagocyte differentiation, polarization, and inflammatory modulation. However, its role is highly context-dependent, exhibiting both pro- and anti-inflammatory properties based on environmental signals, cellular states, and the invading pathogen. KLF4 influences monocyte-to-macrophage differentiation and shapes macrophage polarization, promoting either inflammatory or regulatory phenotypes depending on external cues. In neutrophils, it affects reactive oxygen species production and immune activation, while in dendritic cells, it regulates monocyte-to-dendritic cell differentiation and cytokine secretion. Its diverse involvements in immune responses suggests that it contributes to maintaining a balance between effective pathogen defense and the prevention of excessive and potentially harmful inflammation. This review summarizes current knowledge on the function of KLF4 in phagocytes during infections, highlighting its regulatory mechanisms, context-dependent roles, and its impact on immune activation and resolution. Additionally, potential implications for therapeutic interventions targeting KLF4 are discussed.
Collapse
Affiliation(s)
- Toni Herta
- Department of Hepatology and Gastroenterology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité –Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin, Germany
| | - Aritra Bhattacharyya
- Department of Respiratory Medicine and Critical Care Medicine with Sleep Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Respiratory Medicine and Critical Care Medicine with Sleep Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Janine Zahlten
- Department of Respiratory Medicine and Critical Care Medicine with Sleep Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
5
|
Rijal R, Gomer RH. Pharmacological inhibition of host pathways enhances macrophage killing of intracellular bacterial pathogens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.06.647500. [PMID: 40291742 PMCID: PMC12026824 DOI: 10.1101/2025.04.06.647500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
After ingestion into macrophage phagosomes, some bacterial pathogens such as Mycobacterium tuberculosis ( Mtb ) evade killing by preventing phagosome acidification and fusion of the phagosome with a lysosome. Mtb accumulates extracellular polyphosphate (polyP), and polyP inhibits macrophage phagosome acidification and bacterial killing. In Dictyostelium discoideum , polyP also inhibits bacterial killing, and we identified some proteins in D. discoideum that polyP requires to suppress the killing of ingested bacteria. Here, we find that pharmacological inhibition of human orthologues of the D. discoideum proteins, including P2Y1 receptors, mammalian Target of Rapamycin (mTOR), and inositol hexakisphosphate kinase, enhances the killing of Mtb , Legionella pneumophila , and Listeria monocytogenes by human macrophages. Mtb inhibits phagosome acidification, expression of the proinflammatory marker CD54, and autophagy, and increases expression of the anti-inflammatory marker CD206. In Mtb -infected macrophages, the polyP-degrading enzyme polyphosphatase (ScPPX) and inhibitors reversed these effects, with ScPPX increasing CD54 expression more in female macrophages compared to male macrophages. In addition, Mtb inhibits proteasome activity, and some, but not all, inhibitors reversed these effects. While the existence of a dedicated polyP signaling pathway remains uncertain, our findings suggest that pharmacological inhibition of select host proteins can restore macrophage function and enhances the killing of intracellular pathogens. Importance Human macrophages engulf bacteria into phagosomes, which then fuse with lysosomes to kill the bacteria. However, after engulfment, pathogenic bacteria such as Mycobacterium tuberculosis , Legionella pneumophila , and Listeria monocytogenes can block phagosome-lysosome fusion, allowing their survival. Here, we show that pharmacological inhibition of specific macrophage proteins reverses these effects and enhances bacterial killing. These findings suggest that targeting host factors involved in these processes may provide a therapeutic strategy to improve macrophage function against infections such as tuberculosis, Legionnaires' disease, and listeriosis.
Collapse
|
6
|
Wang X, Zhou H, Li D, Zhao Z, Peng K, Xu X, Wang JJ, Wang Y, Wang J, Zhang JJ, Wan SS, Shi MQ, Chen J, Ding XG, Ji FH. Molecular Targeting of Intracellular Bacteria by Homotypic Recognizing Nanovesicles for Infected Pneumonia Treatment. Biomater Res 2025; 29:0172. [PMID: 40177029 PMCID: PMC11964281 DOI: 10.34133/bmr.0172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/21/2025] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
Although extensive antibiotic regimens have been implemented to address pathogen-infected pneumonia, existing strategies are constrained in their efficacy against intracellular bacteria, a prominent contributor to antibiotic resistance. In addition, the concurrent occurrence of a cytokine storm during antibiotic therapy presents a formidable obstacle in the management of pneumonia caused by pathogens. In the present study, an infection-targeting system that leverages M2-macrophage-derived vesicles [exosomes (Exos)] as vehicles to convey antibiotics (antibiotics@Exos) was developed for effective pneumonia management. The proposed system can enable antibiotics to be specifically delivered to infected macrophages in pneumonia through homotypic recognition and was found to exhibit an exceptional intracellular bactericidal effect. Moreover, M2-type vesicles exhibit a high degree of efficiency in reprogramming inflammatory macrophages toward an anti-inflammatory phenotype. As a result, the administration of antibiotics@Exos was found to substantical decrease the level of the infiltrated inflammatory cells and alleviate the inflammatory factor storm in the lungs of acute lung injury mice. This intervention resulted in the alleviation of reactive-oxygen-species-induced damage, reduction of pulmonary edema, and successful pneumonia treatment. This bioactive vesicle delivery system effectively compensates for the limitations of traditional antibiotic therapy regimens with pluralism effects, paving a new strategy for serious infectious diseases, especially acute pneumonia treatment.
Collapse
Affiliation(s)
- Xu Wang
- Department of Anesthesiology,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hao Zhou
- Department of General Surgery,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu , China
| | - Dan Li
- Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Zhe Zhao
- Suzhou Institute of Nano-Tech and Nano-Bionics, CAS Key Laboratory of Nano-Bio Interface Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - Ke Peng
- Department of Anesthesiology & Institute of Anesthesiology,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiang Xu
- Department of Neurosurgery & Brain and Nerve Research Laboratory,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jia-Jia Wang
- Department of Pulmonary and Critical Care Medicine,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yang Wang
- Department of Anesthesiology,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jun Wang
- Department of Intensive Care Medicine,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jing-Jing Zhang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors,
Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, China
| | - Shuang-Shuang Wan
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors,
Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, China
| | - Mai-Qing Shi
- Department of Anesthesiology,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jun Chen
- Department of Anesthesiology,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xian-Guang Ding
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors,
Nanjing University of Posts and Telecommunications, Nanjing, Jiangsu 210023, China
| | - Fu-Hai Ji
- Department of Anesthesiology & Institute of Anesthesiology,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
7
|
Ubilla-Rodriguez NC, Andreas MP, Giessen TW. Structural and Biochemical Characterization of a Widespread Enterobacterial Peroxidase Encapsulin. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2415827. [PMID: 40167211 DOI: 10.1002/advs.202415827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/11/2025] [Indexed: 04/02/2025]
Abstract
Encapsulins are self-assembling protein compartments found in prokaryotes and specifically encapsulate dedicated cargo enzymes. The most abundant encapsulin cargo class are Dye-decolorizing Peroxidases (DyPs). It has been previously suggested that DyP encapsulins are involved in oxidative stress resistance and bacterial pathogenicity due to DyPs' inherent ability to reduce and detoxify hydrogen peroxide while oxidizing a broad range of organic co-substrates. Here, we report the structural and biochemical analysis of a DyP encapsulin widely found across enterobacteria. Using bioinformatic approaches, we show that this DyP encapsulin is encoded by a conserved transposon-associated operon, enriched in enterobacterial pathogens. Through low pH and peroxide exposure experiments, we highlight the stability of this DyP encapsulin under harsh conditions and show that DyP catalytic activity is highest at low pH. We determine the structure of the DyP-loaded shell and free DyP via cryo-electron microscopy, revealing the structural basis for DyP cargo loading and peroxide preference. This work lays the foundation to further explore the substrate range and physiological functions of enterobacterial DyP encapsulins.
Collapse
Affiliation(s)
| | - Michael P Andreas
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Tobias W Giessen
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| |
Collapse
|
8
|
Xie W, Luo D, Soni V, Wang Z. Functional characterization of MMAR_1296 in Mycobacterium marinum and its potential as a vaccine candidate. Vaccine 2025; 48:126720. [PMID: 39809090 DOI: 10.1016/j.vaccine.2025.126720] [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: 10/08/2024] [Revised: 12/27/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
The Pro-Glu/Pro-Pro-Glu (PE/PPE) family proteins in mycobacteria plays a crucial role in pathogenesis and immune evasion. These proteins characterized by unique structures with conserved sequences. This study elucidated the specific immunological functions of MMAR_1296 from marine mycobacterium. Expressing MMAR_1296 in Mycobacterium smegmatis (M. smegmatis) led to significant alterations in bacterial morphology, as well as reduced survival of M. smegmatis under adverse in vitro conditions and within macrophages. Furthermore, transcriptome analysis of mouse macrophages indicated that natural immunity-related pathways were upregulated in the group infected with M. smegmatis recombinantly expressing MMAR_1296. Moreover, the mycobacterium Growth Inhibition Assays(MGIA)in mice demonstrated that M. smegmatis expressing MMAR_1296 exerted a significant inhibitory effect against Mycobacterium abscessus (M. abscessus) and Mycobacterium marinum (M. marinum) infections. Immunization challenge experiments in mice further confirmed its protective effects, showing a reduction in organ bacterial loads by 1 log10 value compared to the positive control group. These findings indicate that MMAR_1296 is a promising vaccine candidate for M. abscessus and M. marinum. Given that PE/PPE protein family is also a crucial component of Mycobacterium tuberculosis (M. tuberculosis) antigens, further exploration of sequence functions based on MMAR_1296 could reveal broader applications of PE/PPE proteins family for M. tuberculosis treatment. This study supported vaccine development targeting PE/PPE proteins in mycobacteria and paves the way for broader applications.
Collapse
Affiliation(s)
- Weile Xie
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; Collaborative Innovation Center of Agri-Seeds / School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dan Luo
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; Collaborative Innovation Center of Agri-Seeds / School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Vijay Soni
- Division of Infectious Diseases, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Zhe Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; Collaborative Innovation Center of Agri-Seeds / School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
9
|
Liu X, Xu W, Feng J, Wang Y, Li K, Chen Y, Wang W, Zhao W, Ge S, Li J. Adoptive cell transfer of piezo-activated macrophage rescues immunosuppressed rodents from life-threating bacterial infections. Nat Commun 2025; 16:1363. [PMID: 39905015 PMCID: PMC11794888 DOI: 10.1038/s41467-025-56460-2] [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: 02/01/2024] [Accepted: 01/14/2025] [Indexed: 02/06/2025] Open
Abstract
Bacterial infections pose a significant threat to human health. Catalytic antibacterial nanoparticles that generate reactive oxygen species (ROS) are emerging, as a promising therapeutic approach in treating bacterial infection by boosting the innate immune defenses. However, the interaction between innate immune cells and these catalytic nanoparticles remains poorly understood. Here, by using rodent models of bacterial infection, we test the antimicrobial properties of ultrasound-responsive piezo-catalytic nanoparticles (piezoNP). We show that piezoNPs strongly interact with macrophages within subcutaneous abscesses caused by Staphylococcus aureus (S. aureus) infections, and demonstrate that this interaction enhances the macrophage-mediated antibacterial phagocytosis and killing activity through intracellular piezocatalysis. Moreover, we test the use of these piezo-activated macrophages (piezoMϕ) as adoptive cell therapy (ACT) for treating various immunosuppressive bacterial infections, including sepsis, pneumonia and peritonitis. Our study thus highlights the potential application of catalytic nanoparticles as a promising alternative to conventional infection treatment to effectively modulate the innate immune responses and to engineer macrophages for immunotherapy purposes.
Collapse
Affiliation(s)
- Xiaoyi Liu
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Wenxiu Xu
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Junkun Feng
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Ying Wang
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Kai Li
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Yi Chen
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Wenjun Wang
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Weiwei Zhao
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Shaohua Ge
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China.
| | - Jianhua Li
- Department of Biomaterials & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China.
| |
Collapse
|
10
|
Zhou Y, Zhang Y, Li Y, Liu L, Zhuang M, Xiao Y. IL-27 attenuated macrophage injury and inflammation induced by Mycobacterium tuberculosis by activating autophagy. In Vitro Cell Dev Biol Anim 2025; 61:245-256. [PMID: 39455490 DOI: 10.1007/s11626-024-00989-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: 08/14/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
Interleukin-27 (IL-27) is a cytokine that is reported to be highly expressed in the peripheral blood of patients with pulmonary tuberculosis (PTB). IL-27-mediated signaling pathways, which exhibit anti- Mycobacterium tuberculosis (Mtb) properties, have also been demonstrated in macrophages infected with Mtb. However, the exact mechanism remains unclear. This study aimed to clarify the potential molecular mechanisms through which IL-27 enhances macrophage resistance to Mtb infection. Both normal and PTB patients provided bronchoalveolar lavage fluid (BALF). Peripheral blood mononuclear cells (PBMCs) were isolated from healthy individuals and stimulated with 50 ng/mL macrophage-colony stimulating factor (M-CSF) to obtain monocyte-derived macrophages (MDMs). Using 100 ng/mL phorbol 12-myristate 13-acetate (PMA), THP-1 cells were induced to differentiate into THP-1-derived macrophage-like cells (TDMs). Both MDMs and TDMs were subsequently infected with the Mtb strain H37Rv and treated with 50 ng/mL IL-27 prior to infection. The damage and inflammation of macrophages were examined using flow cytometry, enzyme-linked immunosorbent assay (ELISA), and Western blotting. Patients with PTB had elevated levels of IL-27 in their BALF. Preconditioning with IL-27 was shown to reduce H37Rv-induced MDMs and TDMs apoptosis while also decreasing the levels of Cleaved Caspase-3, Bax and the proinflammatory cytokines TNF-α, IL-1β, and IL-6, promoting the expression of Bcl-2 and the anti-inflammatory factors IL-10 and IL-4. Silencing of the IL-27 receptor IL-27Ra increased macrophage damage and inflammation triggered by H37Rv. Mechanistically, IL-27 activates autophagy by inhibiting TLR4/NF-κB signaling and activating the PI3K/AKT signaling pathway, thereby inhibiting H37Rv-induced macrophage apoptosis and the inflammatory response. Our study suggests that IL-27 alleviates H37Rv-induced macrophage injury and the inflammatory response by activating autophagy and that IL-27 may be a new target for the treatment of PTB.
Collapse
Affiliation(s)
- Yushan Zhou
- Respiratory and Critical Care Medicine Ward 1, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, China
| | - Yuxuan Zhang
- Respiratory and Critical Care Medicine Ward 1, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, China
| | - Yanli Li
- Respiratory and Critical Care Medicine Ward 1, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, China
| | - Liqiong Liu
- Respiratory and Critical Care Medicine Ward 1, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, China
| | - Min Zhuang
- Respiratory and Critical Care Medicine Ward 1, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, China
| | - Yi Xiao
- Respiratory and Critical Care Medicine Ward 1, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650051, Yunnan, China.
| |
Collapse
|
11
|
Kooij N, King-Heiden TC. Adaptation of the in vivo respiratory burst assay for fathead minnow larvae (Pimephales promelas). J Immunol Methods 2025; 536:113797. [PMID: 39694456 DOI: 10.1016/j.jim.2024.113797] [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: 05/15/2024] [Revised: 10/25/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Initial innate immune responses such as the respiratory burst response of phagocytes present the first line of defense in response to exposure to pathogens. Several respiratory burst assays have been developed in mammals, cell cultures, and whole zebrafish embryos as a reliable indicator of the innate immune response of a host, and these assays are being used to screen various environmental contaminants for their immunotoxic potential. While zebrafish are a common laboratory fish used in toxicology studies geared towards human health effects, fathead minnows are commonly used as an ecotoxicological indicator species for North America. In this technical note, we describe how we adapted the zebrafish in vivo respiratory burst assay for use in fathead minnow larvae. This assay provides promising expansion of using in vivo respiratory burst responses in different species of larval fish for future comparative immunotoxicity assays, as well as laying the groundwork for studies that can better define the development of the innate and adaptive immune responses of fathead minnow larvae.
Collapse
Affiliation(s)
- Nicole Kooij
- Department of Biological Sciences, University of Wisconsin, La Crosse, United States of America
| | - Tisha C King-Heiden
- Department of Biological Sciences, University of Wisconsin, La Crosse, United States of America.
| |
Collapse
|
12
|
Kotliarova MS, Shumkov MS, Goncharenko AV. Toward Mycobacterium tuberculosis Virulence Inhibition: Beyond Cell Wall. Microorganisms 2024; 13:21. [PMID: 39858789 PMCID: PMC11767696 DOI: 10.3390/microorganisms13010021] [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: 11/20/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/27/2025] Open
Abstract
Mycobacterium tuberculosis (Mtb) is one of the most successful bacterial pathogens in human history. Even in the antibiotic era, Mtb is widespread and causes millions of new cases of tuberculosis each year. The ability to disrupt the host's innate and adaptive immunity, as well as natural persistence, complicates disease control. Tuberculosis traditional therapy involves the long-term use of several antibiotics. Treatment failures are often associated with the development of resistance to one or more drugs. The development of medicines that act on new targets will expand treatment options for tuberculosis caused by multidrug-resistant or extensively drug-resistant Mtb. Therefore, the development of drugs that target virulence factors is an attractive strategy. Such medicines do not have a direct bacteriostatic or bactericidal effect, but can disarm the pathogen so that the host immune system becomes able to eliminate it. Although cell wall-associated targets are being actively studied for anti-TB drug development, other virulence factors important for adaptation and host interaction are also worth comprehensive analysis. In this review, specific Mtb virulence factors (such as secreted phosphatases, regulatory systems, and the ESX-1 secretion system) are identified as promising targets for novel anti-virulence drug development. Additionally, models for the search of virulence inhibitors are discussed, such as virtual screening in silico, in vitro enzyme inhibition assay, the use of recombinant Mtb strains with reporter constructs, phenotypic analysis using in vitro cell infection models and specific environments.
Collapse
Affiliation(s)
- Maria S. Kotliarova
- Bach Institute of Biochemistry, Fundamentals of Biotechnology, Federal Research Center, Russian Academy of Sciences, Moscow 119071, Russia; (M.S.S.); (A.V.G.)
| | | | | |
Collapse
|
13
|
Guo BY, Qi YT, Wu HQ, Zha RY, Wang LJ, Zhang XW, Huang WH. Nanosensor quantitative monitoring of ROS/RNS homeostasis in single phagolysosomes of macrophages during bactericidal processes. Chem Commun (Camb) 2024; 61:97-100. [PMID: 39629638 DOI: 10.1039/d4cc05423g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Reactive oxygen and nitrogen species (ROS/RNS) in macrophages have a potent killing effect on pathogens that infect the host. Here, we achieved in situ, quantitative detection of the homeostasis of four primary ROS/RNS (ONOO-, H2O2, NO, and NO2-) and their precursors (O2˙-, NO) in phagolysosomes of single RAW 264.7 macrophages after phagocytosis of Escherichia coli with platinum-black nanoelectrodes. Enhanced bactericidal activity of the macrophages was observed by an increase in the total amount of ROS/RNS as well as the level and proportion of ONOO-, a potent bactericidal species of RNS. Moreover, both the bactericidal process and the steady-state replenishment process were dominated by the production of RNS (NO-based), revealing differences in the enzyme kinetics of the bactericidal process.
Collapse
Affiliation(s)
- Bing-Yi Guo
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
| | - Yu-Ting Qi
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
| | - Hui-Qian Wu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
| | - Ru-Yan Zha
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
| | - Li-Jun Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
| | - Xin-Wei Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
| | - Wei-Hua Huang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China.
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
14
|
Zhang F, Xia Y, Su J, Quan F, Zhou H, Li Q, Feng Q, Lin C, Wang D, Jiang Z. Neutrophil diversity and function in health and disease. Signal Transduct Target Ther 2024; 9:343. [PMID: 39638788 PMCID: PMC11627463 DOI: 10.1038/s41392-024-02049-y] [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: 05/17/2024] [Revised: 09/21/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Neutrophils, the most abundant type of granulocyte, are widely recognized as one of the pivotal contributors to the acute inflammatory response. Initially, neutrophils were considered the mobile infantry of the innate immune system, tasked with the immediate response to invading pathogens. However, recent studies have demonstrated that neutrophils are versatile cells, capable of regulating various biological processes and impacting both human health and disease. Cytokines and other active mediators regulate the functional activity of neutrophils by activating multiple receptors on these cells, thereby initiating downstream signal transduction pathways. Dysfunctions in neutrophils and disruptions in neutrophil homeostasis have been implicated in the pathogenesis of numerous diseases, including cancer and inflammatory disorders, often due to aberrant intracellular signaling. This review provides a comprehensive synthesis of neutrophil biological functions, integrating recent advancements in this field. Moreover, it examines the biological roles of receptors on neutrophils and downstream signaling pathways involved in the regulation of neutrophil activity. The pathophysiology of neutrophils in numerous human diseases and emerging therapeutic approaches targeting them are also elaborated. This review also addresses the current limitations within the field of neutrophil research, highlighting critical gaps in knowledge that warrant further investigation. In summary, this review seeks to establish a comprehensive and multidimensional model of neutrophil regulation, providing new perspectives for potential clinical applications and further research.
Collapse
Affiliation(s)
- Fengyuan Zhang
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yidan Xia
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jiayang Su
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Fushi Quan
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Hengzong Zhou
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Qirong Li
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Qiang Feng
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Chao Lin
- School of Grain Science and Technology, Jilin Business and Technology College, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China.
| | - Ziping Jiang
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China.
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
15
|
Ubilla-Rodriguez NC, Andreas MP, Giessen TW. Structural and biochemical characterization of a widespread enterobacterial peroxidase encapsulin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.27.625667. [PMID: 39651212 PMCID: PMC11623594 DOI: 10.1101/2024.11.27.625667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Encapsulins are self-assembling protein compartments found in prokaryotes and specifically encapsulate dedicated cargo enzymes. The most abundant encapsulin cargo class are Dye-decolorizing Peroxidases (DyPs). It has been previously suggested that DyP encapsulins are involved in oxidative stress resistance and bacterial pathogenicity due to DyPs' inherent ability to reduce and detoxify hydrogen peroxide while oxidizing a broad range of organic co-substrates. Here, we report the structural and biochemical analysis of a DyP encapsulin widely found across enterobacteria. Using bioinformatic approaches, we show that this DyP encapsulin is encoded by a conserved transposon-associated operon, enriched in enterobacterial pathogens. Through low pH and peroxide exposure experiments, we highlight the stability of this DyP encapsulin under harsh conditions and show that DyP catalytic activity is highest at low pH. We determine the structure of the DyP-loaded shell and free DyP via cryo-electron microscopy, revealing the structural basis for DyP cargo loading and peroxide preference. Our work lays the foundation to further explore the substrate range and physiological functions of enterobacterial DyP encapsulins.
Collapse
|
16
|
Jiang Z, Zhen J, Abulikena Y, Gao C, Huang L, Huang T, Xie J. Mycobacterium tuberculosis VII secretion system effector molecule Rv2347c blocks the maturation of phagosomes and activates the STING/TBK1 signaling pathway to inhibit cell autophagy. Microbiol Spectr 2024; 12:e0118824. [PMID: 39313213 PMCID: PMC11537087 DOI: 10.1128/spectrum.01188-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/01/2024] [Indexed: 09/25/2024] Open
Abstract
The VII secretion system is the main channel for Mycobacterium tuberculosis (MTB) to secrete virulence proteins. The ESAT-like proteins EsxA/B and EsxW/V in the RD region of its genome have been used as targets for vaccine antigens. However, the function of EsxO/P has not been explored, although it was predicted to potentially induce Th1 cell responses as a vaccine development target. In this study, the VII secretion system effector molecule Rv2347c was heterologously expressed in Mycobacterium smegmatis and found to inhibit the expression of the early marker RAB5 of phagosomes, thus preventing the maturation process of phagosomes toward lysosomes, and activated the host cytoplasmic sensing pathway. It inhibited autophagy and activated IFNβ transcription through the STING/TBK1 pathway promoting the host's survival. Therefore, Rv2347c plays an important role in the pathogenesis of MTB with the potential to be utilized as a new target for tuberculosis vaccine development. IMPORTANCE We found that the ESAT-like protein Rv2347c (EsxP) can inhibit the maturation of phagosomes, leading to mycobacterium escape from phagosomes into the cytoplasm, which triggers the host's cytoplasmic sensing pathway STING/TBK1, inhibiting autophagy and upregulating IFNβ transcription, which contributes to the survival of mycobacterium in the host cell. We also found that Rv2347c was able to activate host immunity by activating NF-κB via STING and promoting the transcription of downstream pro-inflammatory factors. Meanwhile, the host also produces IL-1β to repair phagosome maturation arrest via the STING-mediated non-NF-κB pathway.
Collapse
Affiliation(s)
- Zhiyong Jiang
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Junfeng Zhen
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Yuerigu Abulikena
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Chaoyun Gao
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Lingxi Huang
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Tingting Huang
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Jianping Xie
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| |
Collapse
|
17
|
Na-Phatthalung P, Sun S, Xie E, Wang J, Min J, Wang F. The zinc transporter Slc30a1 (ZnT1) in macrophages plays a protective role against attenuated Salmonella. eLife 2024; 13:e89509. [PMID: 39475776 PMCID: PMC11524588 DOI: 10.7554/elife.89509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/05/2024] [Indexed: 11/02/2024] Open
Abstract
The zinc transporter Slc30a1 plays an essential role in maintaining cellular zinc homeostasis. Despite this, its functional role in macrophages remains largely unknown. Here, we examine the function of Slc30a1 in host defense using mice models infected with an attenuated stain of Salmonella enterica Typhimurium and primary macrophages infected with the attenuated Salmonella. Bulk transcriptome sequencing in primary macrophages identifies Slc30a1 as a candidate in response to Salmonella infection. Whole-mount immunofluorescence and confocal microscopy imaging of primary macrophage and spleen from Salmonella-infected Slc30a1flag-EGFP mice demonstrate Slc30a1 expression is increased in infected macrophages with localization at the plasma membrane and in the cytosol. Lyz2-Cre-driven Slc30a1 conditional knockout mice (Slc30a1fl/fl;Lyz2-Cre) exhibit increased susceptibility to Salmonella infection compared to control littermates. We demonstrate that Slc30a1-deficient macrophages are defective in intracellular killing, which correlated with reduced activation of nuclear factor kappa B and reduction in nitric oxide (NO) production. Notably, the model exhibits intracellular zinc accumulation, demonstrating that Slc30a1 is required for zinc export. We thus conclude that zinc export enables the efficient NO-mediated antibacterial activity of macrophages to control invading Salmonella.
Collapse
Affiliation(s)
- Pinanong Na-Phatthalung
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of MedicineHangzhouChina
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of MedicineHangzhouChina
| | - Shumin Sun
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of MedicineHangzhouChina
| | - Enjun Xie
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of MedicineHangzhouChina
| | - Jia Wang
- School of Public Health, Zhengzhou UniversityZhengzhouChina
| | - Junxia Min
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of MedicineHangzhouChina
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of MedicineHangzhouChina
| |
Collapse
|
18
|
Sivaloganathan DM, Wan X, Leon G, Brynildsen MP. Loss of Gre factors leads to phenotypic heterogeneity and cheating in Escherichia coli populations under nitric oxide stress. mBio 2024; 15:e0222924. [PMID: 39248572 PMCID: PMC11498084 DOI: 10.1128/mbio.02229-24] [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: 07/23/2024] [Accepted: 08/05/2024] [Indexed: 09/10/2024] Open
Abstract
Nitric oxide (·NO) is one of the toxic metabolites that bacteria can be exposed to within phagosomes. Gre factors, which are also known as transcript cleavage factors or transcription elongation factors, relieve back-tracked transcription elongation complexes by cleaving nascent RNAs, which allows transcription to resume after stalling. Here we discovered that loss of both Gre factors in Escherichia coli, GreA and GreB, significantly compromised ·NO detoxification due to ·NO-induced phenotypic heterogeneity in ΔgreAΔgreB populations, which did not occur in wild-type cultures. Under normal culturing conditions, both wild-type and ΔgreAΔgreB synthesized transcripts uniformly, whereas treatment with ·NO led to bimodal transcript levels in ΔgreAΔgreB that were unimodal in wild-type. Interestingly, exposure to another toxic metabolite of phagosomes, hydrogen peroxide (H2O2), produced analogous results. Furthermore, we showed that loss of Gre factors led to cheating under ·NO stress where transcriptionally deficient cells benefited from the detoxification activities of the transcriptionally proficient subpopulation. Collectively, these results show that loss of Gre factor activities produces phenotypic heterogeneity under ·NO and H2O2 stress that can yield cheating between subpopulations.IMPORTANCEToxic metabolite stress occurs in a broad range of contexts that are important to human health, microbial ecology, and biotechnology, whereas Gre factors are highly conserved throughout the bacterial kingdom. Here we discovered that loss of Gre factors in E. coli leads to phenotypic heterogeneity under ·NO and H2O2 stress, which we further show with ·NO results in cheating between subpopulations. Collectively, these data suggest that Gre factors play a role in coping with toxic metabolite stress, and that loss of Gre factors can produce cheating between neighbors.
Collapse
Affiliation(s)
| | - Xuanqing Wan
- Department of Chemical
and Biological Engineering, Princeton
University, Princeton,
New Jersey, USA
| | - Gabrielle Leon
- Department of Chemical
and Biological Engineering, Princeton
University, Princeton,
New Jersey, USA
| | - Mark P. Brynildsen
- Department of Chemical
and Biological Engineering, Princeton
University, Princeton,
New Jersey, USA
| |
Collapse
|
19
|
Darwitz BP, Genito CJ, Thurlow LR. Triple threat: how diabetes results in worsened bacterial infections. Infect Immun 2024; 92:e0050923. [PMID: 38526063 PMCID: PMC11385445 DOI: 10.1128/iai.00509-23] [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] [Indexed: 03/26/2024] Open
Abstract
Diabetes mellitus, characterized by impaired insulin signaling, is associated with increased incidence and severity of infections. Various diabetes-related complications contribute to exacerbated bacterial infections, including hyperglycemia, innate immune cell dysfunction, and infection with antibiotic-resistant bacterial strains. One defining symptom of diabetes is hyperglycemia, resulting in elevated blood and tissue glucose concentrations. Glucose is the preferred carbon source of several bacterial pathogens, and hyperglycemia escalates bacterial growth and virulence. Hyperglycemia promotes specific mechanisms of bacterial virulence known to contribute to infection chronicity, including tissue adherence and biofilm formation. Foot infections are a significant source of morbidity in individuals with diabetes and consist of biofilm-associated polymicrobial communities. Bacteria perform complex interspecies behaviors conducive to their growth and virulence within biofilms, including metabolic cross-feeding and altered phenotypes more tolerant to antibiotic therapeutics. Moreover, the metabolic dysfunction caused by diabetes compromises immune cell function, resulting in immune suppression. Impaired insulin signaling induces aberrations in phagocytic cells, which are crucial mediators for controlling and resolving bacterial infections. These aberrancies encompass altered cytokine profiles, the migratory and chemotactic mechanisms of neutrophils, and the metabolic reprogramming required for the oxidative burst and subsequent generation of bactericidal free radicals. Furthermore, the immune suppression caused by diabetes and the polymicrobial nature of the diabetic infection microenvironment may promote the emergence of novel strains of multidrug-resistant bacterial pathogens. This review focuses on the "triple threat" linked to worsened bacterial infections in individuals with diabetes: (i) altered nutritional availability in diabetic tissues, (ii) diabetes-associated immune suppression, and (iii) antibiotic treatment failure.
Collapse
Affiliation(s)
- Benjamin P. Darwitz
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Christopher J. Genito
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill Adams School of Dentistry, Chapel Hill, North Carolina, USA
| | - Lance R. Thurlow
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill Adams School of Dentistry, Chapel Hill, North Carolina, USA
| |
Collapse
|
20
|
Allsup BL, Gharpure S, Bryson BD. Proximity labeling defines the phagosome lumen proteome of murine and primary human macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611277. [PMID: 39282337 PMCID: PMC11398489 DOI: 10.1101/2024.09.04.611277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Proteomic analyses of the phagosome has significantly improved our understanding of the proteins which contribute to critical phagosome functions such as apoptotic cell clearance and microbial killing. However, previous methods of isolating phagosomes for proteomic analysis have relied on cell fractionation with some intrinsic limitations. Here, we present an alternative and modular proximity-labeling based strategy for mass spectrometry proteomic analysis of the phagosome lumen, termed PhagoID. We optimize proximity labeling in the phagosome and apply PhagoID to immortalized murine macrophages as well as primary human macrophages. Analysis of proteins detected by PhagoID in murine macrophages demonstrate that PhagoID corroborates previous proteomic studies, but also nominates novel proteins with unexpected residence at the phagosome for further study. A direct comparison between the proteins detected by PhagoID between mouse and human macrophages further reveals that human macrophage phagosomes have an increased abundance of proteins involved in the oxidative burst and antigen presentation. Our study develops and benchmarks a new approach to measure the protein composition of the phagosome and validates a subset of these findings, ultimately using PhagoID to grant further insight into the core constituent proteins and species differences at the phagosome lumen.
Collapse
Affiliation(s)
- Benjamin L Allsup
- Department of Biological Engineering, MIT, Cambridge, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, USA
| | - Supriya Gharpure
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, USA
| | - Bryan D Bryson
- Department of Biological Engineering, MIT, Cambridge, USA
- Ragon Institute of Mass General, Harvard, and MIT, Cambridge, USA
| |
Collapse
|
21
|
Han XQ, Cui ZW, Ma ZY, Wang J, Hu YZ, Li J, Ye JM, Tafalla C, Zhang YA, Zhang XJ. Phagocytic Plasma Cells in Teleost Fish Provide Insights into the Origin and Evolution of B Cells in Vertebrates. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:730-742. [PMID: 38984862 DOI: 10.4049/jimmunol.2400182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 06/17/2024] [Indexed: 07/11/2024]
Abstract
Teleost IgM+ B cells can phagocytose, like mammalian B1 cells, and secrete Ag-specific IgM, like mammalian B2 cells. Therefore, teleost IgM+ B cells may have the functions of both mammalian B1 and B2 cells. To support this view, we initially found that grass carp (Ctenopharyngodon idella) IgM+ plasma cells (PCs) exhibit robust phagocytic ability, akin to IgM+ naive B cells. Subsequently, we sorted grass carp IgM+ PCs into two subpopulations: nonphagocytic (Pha-IgM+ PCs) and phagocytic IgM+ PCs (Pha+IgM+ PCs), both of which demonstrated the capacity to secrete natural IgM with LPS and peptidoglycan binding capacity. Remarkably, following immunization of grass carp with an Ag, we observed that both Pha-IgM+ PCs and Pha+IgM+ PCs could secrete Ag-specific IgM. Furthermore, in vitro concatenated phagocytosis experiments in which Pha-IgM+ PCs from an initial phagocytosis experiment were sorted and exposed again to beads confirmed that these cells also have phagocytic capabilities, thereby suggesting that all teleost IgM+ B cells have phagocytic potential. Additionally, we found that grass carp IgM+ PCs display classical phenotypic features of macrophages, providing support for the hypothesis that vertebrate B cells evolved from ancient phagocytes. These findings together reveal that teleost B cells are a primitive B cell type with functions reminiscent of both mammalian B1 and B2 cells, providing insights into the origin and evolution of B cells in vertebrates.
Collapse
Affiliation(s)
- Xue-Qing Han
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Zheng-Wei Cui
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Zi-You Ma
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Jie Wang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Ya-Zhen Hu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Jun Li
- School of Science and Medicine, Lake Superior State University, Sault Sainte Marie, MI
| | - Jian-Min Ye
- Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, Guangdong Provincial Engineering Technology Research Center for Environmentally-Friendly Aquaculture, Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Institute of Modern Aquaculture Science and Engineering, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Carolina Tafalla
- Animal Health Research Center (CISA), National Institute for Agricultural and Food Research and Technology (INIA), National Research Council (CSIC), Madrid, Spain
| | - Yong-An Zhang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Xu-Jie Zhang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| |
Collapse
|
22
|
Di T, Chen Y, Zhou Z, Liu J, Du Y, Feng C, Zhu B, Wang L. Effect of α7 nAChR-autophagy axis of deciduous tooth pulp stem cells in regulating IL-1β in the process of physiological root resorption of deciduous teeth. J Mol Med (Berl) 2024; 102:1135-1149. [PMID: 39002004 DOI: 10.1007/s00109-024-02466-0] [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: 12/31/2023] [Revised: 05/27/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
Physiological root resorption of deciduous teeth is a normal phenomenon occurring during the developmental stages of children. Previous research has indicated the pivotal role of the inflammatory microenvironment in this process, although the specific mechanisms remain unclear. This study is aimed at elucidating the involvement of the alpha7 nicotinic acetylcholine receptors (α7 nAChR)-autophagy axis in the regulation of the inflammatory microenvironment during physiological root resorption in deciduous teeth. Samples were collected from deciduous teeth at various stages of physiological root resorption, and deciduous dental pulp stem cells (DDPSCs) were isolated and cultured during the mid-phase of root resorption. The findings revealed a substantial infiltration of the pulp of deciduous teeth at the mid-phase of root resorption, characterized by elevated expression levels of α7 nAChR and IL-1β. Significantly increased IL-1β and α7 nAChR expressions were observed in DDPSCs during the mid-phase of root resorption, with α7 nAChR demonstrating a regulatory effect on IL-1β. Moreover, evidence suggested that mechanical stress may act as a trigger, regulating autophagy and IL-1 expression via α7 nAChR. In conclusion, mechanical stress was identified as a regulator of autophagy in DDPSCs through α7 nAChR, influencing the expression of IL-1β and contributing to the formation of the inflammatory microenvironment. This mechanism plays a crucial role in the physiological root resorption of deciduous teeth. KEY MESSAGES: The pulp of deciduous teeth at mid-phase of root resorption was heavily infiltrated with high expression of α7nAChR and IL-1β. α7 nAChR acts as an initiating factor to regulate IL-1β through autophagy in DDPSCs. Mechanical stress can regulate autophagy of DDPSCs through α7 nAChR and thus affect IL-1β expression and inflammatory microenvironment formation in physiological root resorption in deciduous teeth.
Collapse
Affiliation(s)
- Tiankai Di
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
- Department of Stomatology, The 969th Hospital, Joint Logistics Support Force of the Chinese People's Liberation Army, Hohhot, Inner Mongolia, 010000, People's Republic of China
| | - Yujiang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Zhifei Zhou
- Department of Stomatology, General Hospital of Tibet Military Region, Lhasa, 850007, People's Republic of China
| | - Jiajia Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Yang Du
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China
| | - Chao Feng
- Department of Stomatology, The 969th Hospital, Joint Logistics Support Force of the Chinese People's Liberation Army, Hohhot, Inner Mongolia, 010000, People's Republic of China
- Center for Computational Biology, Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing, 100850, People's Republic of China
| | - Bin Zhu
- Department of Stomatology, General Hospital of Tibet Military Region, Lhasa, 850007, People's Republic of China.
| | - Lulu Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, People's Republic of China.
| |
Collapse
|
23
|
Bellucci MC, Romani C, Sani M, Volonterio A. Dual Antibiotic Approach: Synthesis and Antibacterial Activity of Antibiotic-Antimicrobial Peptide Conjugates. Antibiotics (Basel) 2024; 13:783. [PMID: 39200083 PMCID: PMC11352213 DOI: 10.3390/antibiotics13080783] [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: 07/23/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/01/2024] Open
Abstract
In recent years, bacterial resistance to conventional antibiotics has become a major concern in the medical field. The global misuse of antibiotics in clinics, personal use, and agriculture has accelerated this resistance, making infections increasingly difficult to treat and rendering new antibiotics ineffective more quickly. Finding new antibiotics is challenging due to the complexity of bacterial mechanisms, high costs and low financial incentives for the development of new molecular scaffolds, and stringent regulatory requirements. Additionally, innovation has slowed, with many new antibiotics being modifications of existing drugs rather than entirely new classes. Antimicrobial peptides (AMPs) are a valid alternative to small-molecule antibiotics offering several advantages, including broad-spectrum activity and a lower likelihood of inducing resistance due to their multifaceted mechanisms of action. However, AMPs face challenges such as stability issues in physiological conditions, potential toxicity to human cells, high production costs, and difficulties in large-scale manufacturing. A reliable strategy to overcome the drawbacks associated with the use of small-molecule antibiotics and AMPs is combination therapy, namely the simultaneous co-administration of two or more antibiotics or the synthesis of covalently linked conjugates. This review aims to provide a comprehensive overview of the literature on the development of antibiotic-AMP conjugates, with a particular emphasis on critically analyzing the design and synthetic strategies employed in their creation. In addition to the synthesis, the review will also explore the reported antibacterial activity of these conjugates and, where available, examine any data concerning their cytotoxicity.
Collapse
Affiliation(s)
- Maria Cristina Bellucci
- Department of Food, Environmental, and Nutritional Sciences, Università degli Studi di Milano, Via Celoria 2, 20131 Milano, Italy;
| | - Carola Romani
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy;
| | - Monica Sani
- Consiglio Nazionale delle Ricerche, Istituto di Scienze e Tecnologie Chimica “G. Natta” (SCITEC), Via Mario Bianco 9, 20131 Milano, Italy;
| | - Alessandro Volonterio
- Department of Chemistry, Materials, and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy;
| |
Collapse
|
24
|
Harioudh MK, Perez J, So L, Maheshwari M, Ebert TS, Hornung V, Savan R, Rouf Banday A, Diamond MS, Rathinam VA, Sarkar SN. The canonical antiviral protein oligoadenylate synthetase 1 elicits antibacterial functions by enhancing IRF1 translation. Immunity 2024; 57:1812-1827.e7. [PMID: 38955184 PMCID: PMC11324410 DOI: 10.1016/j.immuni.2024.06.003] [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: 11/21/2023] [Revised: 04/11/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024]
Abstract
An important property of the host innate immune response during microbial infection is its ability to control the expression of antimicrobial effector proteins, but how this occurs post-transcriptionally is not well defined. Here, we describe a critical antibacterial role for the classic antiviral gene 2'-5'-oligoadenylate synthetase 1 (OAS1). Human OAS1 and its mouse ortholog, Oas1b, are induced by interferon-γ and protect against cytosolic bacterial pathogens such as Francisella novicida and Listeria monocytogenes in vitro and in vivo. Proteomic and transcriptomic analysis showed reduced IRF1 protein expression in OAS1-deficient cells. Mechanistically, OAS1 binds and localizes IRF1 mRNA to the rough endoplasmic reticulum (ER)-Golgi endomembranes, licensing effective translation of IRF1 mRNA without affecting its transcription or decay. OAS1-dependent translation of IRF1 leads to the enhanced expression of antibacterial effectors, such as GBPs, which restrict intracellular bacteria. These findings uncover a noncanonical function of OAS1 in antibacterial innate immunity.
Collapse
Affiliation(s)
- Munesh K Harioudh
- Cancer Virology Program, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Joseph Perez
- Cancer Virology Program, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lomon So
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA, USA
| | - Mayank Maheshwari
- Cancer Virology Program, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Thomas S Ebert
- Department of Biochemistry, Ludwig Maximilians Universität, Munich, Germany
| | - Veit Hornung
- Department of Biochemistry, Ludwig Maximilians Universität, Munich, Germany
| | - Ram Savan
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA, USA
| | - A Rouf Banday
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Vijay A Rathinam
- Department of Immunology, UConn Health School of Medicine, Farmington, CT, USA
| | - Saumendra N Sarkar
- Cancer Virology Program, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
25
|
Wang W, Mo W, Xiao X, Cai M, Feng S, Wang Y, Zhou D. Antibiotic-loaded lactoferrin nanoparticles as a platform for enhanced infection therapy through targeted elimination of intracellular bacteria. Asian J Pharm Sci 2024; 19:100926. [PMID: 39253610 PMCID: PMC11381595 DOI: 10.1016/j.ajps.2024.100926] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/01/2024] [Accepted: 04/02/2024] [Indexed: 09/11/2024] Open
Abstract
Intracellular bacteria can multiply inside host cells and manipulate their biology, and the efficacy of traditional antibiotic drug therapy for intracellular bacteria is limited by inadequate drug accumulation. Fighting against these stealthy bacteria has been a long-standing challenge. Here, a system of stimuli-responsive lactoferrin (Lf) nanoparticles is prepared using protein self-assembly technology to deliver broad-spectrum antibiotic rifampicin (Rif) (Rif@Lf NPs) for enhanced infection therapy through targeted elimination of intracellular bacteria. Compared to Rif@BSA NPs, the Rif@Lf NPs can specifically target macrophages infected by bacteria, thus increasing the accumulation of Rif within macrophages. Subsequently, Rif@Lf NPs with positive surface charge further displayed targeted adherence to the bacteria within macrophages and released Rif rapidly in a redox-responsive manner. Combined with the antibacterial activities of Lf and Rif, the Rif@Lf NPs showed broad-spectrum antibiotic abilities to intracellular bacteria and biofilms. As a result, the Rif@Lf NPs with high safety exhibited excellent therapeutic efficacy in the disease models of subcutaneous infection, sepsis, and bacterial keratitis. Taken together, the antibiotic-loaded Lf nanoparticles present a promising platform to combat pathogen infections through targeted elimination of intracellular bacteria.
Collapse
Affiliation(s)
- Wei Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wanying Mo
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xue Xiao
- Department of Ophthalmology & Department of Ultrasonic Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Manying Cai
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Songfu Feng
- Department of Ophthalmology & Department of Ultrasonic Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yupeng Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Department of Ophthalmology & Department of Ultrasonic Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Dongfang Zhou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Department of Ophthalmology & Department of Ultrasonic Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
26
|
Pedersen BH, Simões FB, Pogrebnyakov I, Welch M, Johansen HK, Molin S, La Rosa R. Metabolic specialization drives reduced pathogenicity in Pseudomonas aeruginosa isolates from cystic fibrosis patients. PLoS Biol 2024; 22:e3002781. [PMID: 39178315 PMCID: PMC11376529 DOI: 10.1371/journal.pbio.3002781] [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: 09/05/2024] [Accepted: 08/01/2024] [Indexed: 08/25/2024] Open
Abstract
Metabolism provides the foundation for all cellular functions. During persistent infections, in adapted pathogenic bacteria metabolism functions radically differently compared with more naïve strains. Whether this is simply a necessary accommodation to the persistence phenotype or if metabolism plays a direct role in achieving persistence in the host is still unclear. Here, we characterize a convergent shift in metabolic function(s) linked with the persistence phenotype during Pseudomonas aeruginosa colonization in the airways of people with cystic fibrosis. We show that clinically relevant mutations in the key metabolic enzyme, pyruvate dehydrogenase, lead to a host-specialized metabolism together with a lower virulence and immune response recruitment. These changes in infection phenotype are mediated by impaired type III secretion system activity and by secretion of the antioxidant metabolite, pyruvate, respectively. Our results show how metabolic adaptations directly impinge on persistence and pathogenicity in this organism.
Collapse
Affiliation(s)
- Bjarke Haldrup Pedersen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Filipa Bica Simões
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark
| | - Ivan Pogrebnyakov
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Martin Welch
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Helle Krogh Johansen
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Molin
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Ruggero La Rosa
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
27
|
Ondondo B. Editorial: Overcoming challenges in microbial immunology: 2022. Front Immunol 2024; 15:1436631. [PMID: 38953029 PMCID: PMC11215133 DOI: 10.3389/fimmu.2024.1436631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 06/05/2024] [Indexed: 07/03/2024] Open
Affiliation(s)
- Beatrice Ondondo
- Immunology Department, University Hospitals of Leicester National Health Service (NHS) Trust, Leicester, United Kingdom
| |
Collapse
|
28
|
Sun J, Chen C, Pan P, Zhang K, Xu J, Chen C. The potential of bacterial anti-phagocytic proteins in suppressing the clearance of extracellular vesicles mediated by host phagocytosis. Front Immunol 2024; 15:1418061. [PMID: 38903499 PMCID: PMC11186983 DOI: 10.3389/fimmu.2024.1418061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024] Open
Abstract
Extracellular vesicles (EVs), characterized by low immunogenicity, high biocompatibility and targeting specificity along with excellent blood-brain barrier permeability, are increasingly recognized as promising drug delivery vehicles for treating a variety of diseases, such as cancer, inflammation and viral infection. However, recent findings demonstrate that the intracellular delivery efficiency of EVs fall short of expectations due to phagocytic clearance mediated by the host mononuclear phagocyte system through Fcγ receptors, complement receptors as well as non-opsonic phagocytic receptors. In this text, we investigate a range of bacterial virulence proteins that antagonize host phagocytic machinery, aiming to explore their potential in engineering EVs to counteract phagocytosis. Special emphasis is placed on IdeS secreted by Group A Streptococcus and ImpA secreted by Pseudomonas aeruginosa, as they not only counteract phagocytosis but also bind to highly upregulated surface biomarkers αVβ3 on cancer cells or cleave the tumor growth and metastasis-promoting factor CD44, respectively. This suggests that bacterial anti-phagocytic proteins, after decorated onto EVs using pre-loading or post-loading strategies, can not only improve EV-based drug delivery efficiency by evading host phagocytosis and thus achieve better therapeutic outcomes but also further enable an innovative synergistic EV-based cancer therapy approach by integrating both phagocytosis antagonism and cancer targeting or deactivation.
Collapse
Affiliation(s)
- Jiacong Sun
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Congcong Chen
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Pengpeng Pan
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Keyi Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Jinrui Xu
- School of Life Sciences, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, China
| | - Cheng Chen
- School of Life Sciences, Tianjin University, Tianjin, China
| |
Collapse
|
29
|
Burgos-Molina AM, Téllez Santana T, Redondo M, Bravo Romero MJ. The Crucial Role of Inflammation and the Immune System in Colorectal Cancer Carcinogenesis: A Comprehensive Perspective. Int J Mol Sci 2024; 25:6188. [PMID: 38892375 PMCID: PMC11172443 DOI: 10.3390/ijms25116188] [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: 05/04/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Chronic inflammation drives the growth of colorectal cancer through the dysregulation of molecular pathways within the immune system. Infiltration of immune cells, such as macrophages, into tumoral regions results in the release of proinflammatory cytokines (IL-6; IL-17; TNF-α), fostering tumor proliferation, survival, and invasion. Tumors employ various mechanisms to evade immune surveillance, effectively 'cloaking' themselves from detection and subsequent attack. A comprehensive understanding of these intricate molecular interactions is paramount for advancing novel strategies aimed at modulating the immune response against cancer.
Collapse
Affiliation(s)
- Antonio Manuel Burgos-Molina
- Surgery, Biochemistry and Immunology Department, School of Medicine, University of Malaga, 29010 Málaga, Spain; (A.M.B.-M.); (T.T.S.); (M.J.B.R.)
| | - Teresa Téllez Santana
- Surgery, Biochemistry and Immunology Department, School of Medicine, University of Malaga, 29010 Málaga, Spain; (A.M.B.-M.); (T.T.S.); (M.J.B.R.)
- Research Network on Chronic Diseases, Primary Care, and Health Promotion (RICAPPS), Carlos III Health Institute (Instituto de Salud Carlos III), Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
- Málaga Biomedical Research Institute (Instituto de Investigación Biomédica de Málaga, IBIMA), Calle Doctor Miguel Díaz Recio, 28, 29010 Málaga, Spain
| | - Maximino Redondo
- Surgery, Biochemistry and Immunology Department, School of Medicine, University of Malaga, 29010 Málaga, Spain; (A.M.B.-M.); (T.T.S.); (M.J.B.R.)
- Research Network on Chronic Diseases, Primary Care, and Health Promotion (RICAPPS), Carlos III Health Institute (Instituto de Salud Carlos III), Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
- Málaga Biomedical Research Institute (Instituto de Investigación Biomédica de Málaga, IBIMA), Calle Doctor Miguel Díaz Recio, 28, 29010 Málaga, Spain
- Research Unit, Hospital Costa del Sol, Autovía A-7, km 187, 29603 Marbella, Spain
| | - María José Bravo Romero
- Surgery, Biochemistry and Immunology Department, School of Medicine, University of Malaga, 29010 Málaga, Spain; (A.M.B.-M.); (T.T.S.); (M.J.B.R.)
| |
Collapse
|
30
|
Xu H, Wang X, Zhang Z, Hu J, Yu Y, Wang J, Liu Y, Liu J. Staphylococcus aureus promotes its intracellular survival by inhibiting Rab11-Rab11FIP4-mediated vesicle trafficking. Vet Microbiol 2024; 293:110091. [PMID: 38626624 DOI: 10.1016/j.vetmic.2024.110091] [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: 09/07/2023] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/18/2024]
Abstract
Mastitis in dairy cows is mainly caused by bacteria, in which Staphylococcus aureus appears frequently. Epithelial cells, as a major physical barrier of mammary gland, play an important role in preventing mastitis in dairy cows. Our previous study reported that Rab11fip4 (an effector of Rab11) was significantly changed in response to stimulation by S. aureus. So, in this study, the role of Rab11A in phagocytosis of bovine mammary epithelial cells (MAC-T) against S. aureus was evaluated. First, changes of Rab11A and Rab11fip4 were analyzed in response to S. aureus by immunofluorescence and western blotting. Subsequently, the effects of Rab11A and Rab11fip4 on proliferation of S. aureus, as well as formation and function of late endosomes (LEs) and lysosomes (LYSs) were investigated. The results showed that, after infection, Rab11A and Rab11fip4 were recruited to phagosomes containing S. aureus. Rab11A promoted bacterial clearance and rescues the destruction of LEs and LYSs by S. aureus, whereas Rab11fip4 did the opposite. These findings provide new insights into phagocytosis and control of S. aureus in host cells, thus lay the foundation to elucidate the pathogenesis of S. aureus in bovine mastitis.
Collapse
Affiliation(s)
- Huiling Xu
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong 271018, China
| | - Xiaozhou Wang
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong 271018, China
| | - Zhizhong Zhang
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong 271018, China
| | - Jiaqing Hu
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong 271018, China
| | - Yongtao Yu
- School of Animal Science and Technology, Ningxia University, Yinchuan 750000, China
| | - Jiandong Wang
- Institute of Animal Science, Ningxia Academy of Agricultural and Forestry Sciences, Yinchuan, Ningxia 750002, China
| | - Yongxia Liu
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong 271018, China; Research Center for Animal Disease Control Engineering, Shandong Agricultural University, Tai`an, Shandong 271018, China.
| | - Jianzhu Liu
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, Shandong 271018, China.
| |
Collapse
|
31
|
Peng C, Cheng Y, Ma M, Chen Q, Duan Y, Liu S, Cheng H, Yang H, Huang J, Bu W, Shi C, Wu X, Chen J, Zheng R, Liu Z, Ji Z, Wang J, Huang X, Wang P, Sha W, Ge B, Wang L. Mycobacterium tuberculosis suppresses host antimicrobial peptides by dehydrogenating L-alanine. Nat Commun 2024; 15:4216. [PMID: 38760394 PMCID: PMC11101664 DOI: 10.1038/s41467-024-48588-4] [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: 09/18/2023] [Accepted: 05/07/2024] [Indexed: 05/19/2024] Open
Abstract
Antimicrobial peptides (AMPs), ancient scavengers of bacteria, are very poorly induced in macrophages infected by Mycobacterium tuberculosis (M. tuberculosis), but the underlying mechanism remains unknown. Here, we report that L-alanine interacts with PRSS1 and unfreezes the inhibitory effect of PRSS1 on the activation of NF-κB pathway to induce the expression of AMPs, but mycobacterial alanine dehydrogenase (Ald) Rv2780 hydrolyzes L-alanine and reduces the level of L-alanine in macrophages, thereby suppressing the expression of AMPs to facilitate survival of mycobacteria. Mechanistically, PRSS1 associates with TAK1 and disruptes the formation of TAK1/TAB1 complex to inhibit TAK1-mediated activation of NF-κB pathway, but interaction of L-alanine with PRSS1, disables PRSS1-mediated impairment on TAK1/TAB1 complex formation, thereby triggering the activation of NF-κB pathway to induce expression of AMPs. Moreover, deletion of antimicrobial peptide gene β-defensin 4 (Defb4) impairs the virulence by Rv2780 during infection in mice. Both L-alanine and the Rv2780 inhibitor, GWP-042, exhibits excellent inhibitory activity against M. tuberculosis infection in vivo. Our findings identify a previously unrecognized mechanism that M. tuberculosis uses its own alanine dehydrogenase to suppress host immunity, and provide insights relevant to the development of effective immunomodulators that target M. tuberculosis.
Collapse
Affiliation(s)
- Cheng Peng
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai, China
| | - Yuanna Cheng
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai, China
| | - Mingtong Ma
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai, China
| | - Qiu Chen
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai, China
| | - Yongjia Duan
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai, China
| | - Shanshan Liu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai, China
| | - Hongyu Cheng
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai, China
| | - Hua Yang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingping Huang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai, China
| | - Wenyi Bu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai, China
| | - Chenyue Shi
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai, China
| | - Xiangyang Wu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Clinical Translation Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianxia Chen
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Clinical Translation Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ruijuan Zheng
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhonghua Liu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhe Ji
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai, China
| | - Jie Wang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaochen Huang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Peng Wang
- Shanghai Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei Sha
- Shanghai Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Baoxue Ge
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai, China.
- Shanghai Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
- Clinical Translation Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Lin Wang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai, China.
- Shanghai Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
32
|
Goh KGK, Desai D, Thapa R, Prince D, Acharya D, Sullivan MJ, Ulett GC. An opportunistic pathogen under stress: how Group B Streptococcus responds to cytotoxic reactive species and conditions of metal ion imbalance to survive. FEMS Microbiol Rev 2024; 48:fuae009. [PMID: 38678005 PMCID: PMC11098048 DOI: 10.1093/femsre/fuae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 03/26/2024] [Accepted: 04/16/2024] [Indexed: 04/29/2024] Open
Abstract
Group B Streptococcus (GBS; also known as Streptococcus agalactiae) is an opportunistic bacterial pathogen that causes sepsis, meningitis, pneumonia, and skin and soft tissue infections in neonates and healthy or immunocompromised adults. GBS is well-adapted to survive in humans due to a plethora of virulence mechanisms that afford responses to support bacterial survival in dynamic host environments. These mechanisms and responses include counteraction of cell death from exposure to excess metal ions that can cause mismetallation and cytotoxicity, and strategies to combat molecules such as reactive oxygen and nitrogen species that are generated as part of innate host defence. Cytotoxicity from reactive molecules can stem from damage to proteins, DNA, and membrane lipids, potentially leading to bacterial cell death inside phagocytic cells or within extracellular spaces within the host. Deciphering the ways in which GBS responds to the stress of cytotoxic reactive molecules within the host will benefit the development of novel therapeutic and preventative strategies to manage the burden of GBS disease. This review summarizes knowledge of GBS carriage in humans and the mechanisms used by the bacteria to circumvent killing by these important elements of host immune defence: oxidative stress, nitrosative stress, and stress from metal ion intoxication/mismetallation.
Collapse
Affiliation(s)
- Kelvin G K Goh
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| | - Devika Desai
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| | - Ruby Thapa
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| | - Darren Prince
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| | - Dhruba Acharya
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| | - Matthew J Sullivan
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Glen C Ulett
- School of Pharmacy and Medical Sciences, and Menzies Health Institute Queensland, Griffith University, Parklands Drive, Southport, Gold Coast Campus, QLD 4222, Australia
| |
Collapse
|
33
|
Abstract
The recent emergence of nanomedicine has revolutionized the therapeutic landscape and necessitated the creation of more sophisticated drug delivery systems. Polymeric nanoparticles sit at the forefront of numerous promising drug delivery designs, due to their unmatched control over physiochemical properties such as size, shape, architecture, charge, and surface functionality. Furthermore, polymeric nanoparticles have the ability to navigate various biological barriers to precisely target specific sites within the body, encapsulate a diverse range of therapeutic cargo and efficiently release this cargo in response to internal and external stimuli. However, despite these remarkable advantages, the presence of polymeric nanoparticles in wider clinical application is minimal. This review will provide a comprehensive understanding of polymeric nanoparticles as drug delivery vehicles. The biological barriers affecting drug delivery will be outlined first, followed by a comprehensive description of the various nanoparticle designs and preparation methods, beginning with the polymers on which they are based. The review will meticulously explore the current performance of polymeric nanoparticles against a myriad of diseases including cancer, viral and bacterial infections, before finally evaluating the advantages and crucial challenges that will determine their wider clinical potential in the decades to come.
Collapse
Affiliation(s)
- Maximilian
A. Beach
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Umeka Nayanathara
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yanting Gao
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Changhe Zhang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yijun Xiong
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yufu Wang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Georgina K. Such
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
34
|
Daman AW, Cheong JG, Berneking L, Josefowicz SZ. The potency of hematopoietic stem cell reprogramming for changing immune tone. Immunol Rev 2024; 323:197-208. [PMID: 38632868 DOI: 10.1111/imr.13335] [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] [Indexed: 04/19/2024]
Abstract
Innate immune memory endows innate immune cells with antigen independent heightened responsiveness to subsequent challenges. The durability of this response can be mediated by inflammation induced epigenetic and metabolic reprogramming in hematopoietic stem and progenitor cells (HSPCs) that are maintained through differentiation to mature immune progeny. Understanding the mechanisms and extent of trained immunity induction by pathogens and vaccines, such as BCG, in HSPC remains a critical area of exploration with important implications for health and disease. Here we review these concepts and present new analysis to highlight how inflammatory reprogramming of HSPC can potently alter immune tone, including to enhance specific anti-tumor responses. New findings in the field pave the way for novel HSPC targeting therapeutic strategies in cancer and other contexts of immune modulation. Future studies are expected to unravel diverse and extensive effects of infections, vaccines, microbiota, and sterile inflammation on hematopoietic progenitor cells and begin to illuminate the broad spectrum of immunologic tuning that can be established through altering HSPC phenotypes. The purpose of this review is to draw attention to emerging and speculative topics in this field where we posit that focused study of HSPC in the framework of trained immunity holds significant promise.
Collapse
Affiliation(s)
- Andrew W Daman
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, New York, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Jin Gyu Cheong
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, New York, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Laura Berneking
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Steven Z Josefowicz
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, New York, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
35
|
Ullah H, Shi X, Taj A, Cheng L, Yan Q, Sha S, Ahmad, Kang J, Haris M, Ma X, Ma Y. Mycobacterium tuberculosis PE_PGRS38 Enhances Intracellular Survival of Mycobacteria by Inhibiting TLR4/NF-κB-Dependent Inflammation and Apoptosis of the Host. BIOLOGY 2024; 13:313. [PMID: 38785795 PMCID: PMC11118070 DOI: 10.3390/biology13050313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/20/2024] [Accepted: 04/27/2024] [Indexed: 05/25/2024]
Abstract
Mycobacterium tuberculosis (Mtb) ranks as the most lethal human pathogen, able to fend off repeated attacks by the immune system or medications. PE_PGRS proteins are hallmarks of the pathogenicity of Mtb and contribute to its antigenic diversity, virulence, and persistence during infection. M. smegmatis is a nonpathogenic mycobacterium that naturally lacks PE_PGRS and is used as a model to express Mtb proteins. PE_PGRS has the capability to evade host immune responses and enhance the intracellular survival of M. smegmatis. Despite the intense investigations into PE_PGRS proteins, their role in tuberculosis remains elusive. We engineered the recombinant M. smegmatis strain Ms-PE_PGRS38. The result shows that PE_PGRS38 is expressed in the cell wall of M. smegmatis. PE_PGRS38 contributes to biofilm formation, confers permeability to the cell wall, and shows variable responses to exogenous stresses. PE_PGRS38 downregulated TLR4/NF-κB signaling in RAW264.7 macrophages and lung tissues of infected mice. In addition, PE_PGRS38 decreased NLRP3-dependent IL-1β release and limited pathogen-mediated inflammasome activity during infection. Moreover, PE_PGRS38 inhibited the apoptosis of RAW264.7 cells by downregulating the expression of apoptotic markers including Bax, cytochrome c, caspase-3, and caspase-9. In a nutshell, our findings demonstrate that PE_PGRS38 is a virulence factor for Mtb that enables recombinant M. smegmatis to survive by resisting and evading the host's immune responses during infection.
Collapse
Affiliation(s)
- Hayan Ullah
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (H.U.); (A.T.); (L.C.); (S.S.); (J.K.); (M.H.)
- Department of Microbiology, Dalian Medical University, Dalian 116044, China;
| | - Xiaoxia Shi
- Department of Experimental Teaching Center of Public Health, Dalian Medical University, Dalian 116044, China;
| | - Ayaz Taj
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (H.U.); (A.T.); (L.C.); (S.S.); (J.K.); (M.H.)
| | - Lin Cheng
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (H.U.); (A.T.); (L.C.); (S.S.); (J.K.); (M.H.)
| | - Qiulong Yan
- Department of Microbiology, Dalian Medical University, Dalian 116044, China;
| | - Shanshan Sha
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (H.U.); (A.T.); (L.C.); (S.S.); (J.K.); (M.H.)
| | - Ahmad
- Department of Immunology, Dalian Medical University, Dalian 116044, China;
| | - Jian Kang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (H.U.); (A.T.); (L.C.); (S.S.); (J.K.); (M.H.)
| | - Muhammad Haris
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (H.U.); (A.T.); (L.C.); (S.S.); (J.K.); (M.H.)
| | - Xiaochi Ma
- Pharmaceutical Research Center, The Second Affiliated Hospital, Dalian Medical University, Dalian 116044, China
| | - Yufang Ma
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, China; (H.U.); (A.T.); (L.C.); (S.S.); (J.K.); (M.H.)
- Department of Microbiology, Dalian Medical University, Dalian 116044, China;
| |
Collapse
|
36
|
Pugazhendhi AS, Seal A, Hughes M, Kumar U, Kolanthai E, Wei F, Schwartzman JD, Coathup MJ. Extracellular Proteins Isolated from L. acidophilus as an Osteomicrobiological Therapeutic Agent to Reduce Pathogenic Biofilm Formation, Regulate Chronic Inflammation, and Augment Bone Formation In Vitro. Adv Healthc Mater 2024; 13:e2302835. [PMID: 38117082 DOI: 10.1002/adhm.202302835] [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: 08/25/2023] [Revised: 10/31/2023] [Indexed: 12/21/2023]
Abstract
Periprosthetic joint infection (PJI) is a challenging complication that can occur following joint replacement surgery. Efficacious strategies to prevent and treat PJI and its recurrence remain elusive. Commensal bacteria within the gut convey beneficial effects through a defense strategy named "colonization resistance" thereby preventing pathogenic infection along the intestinal surface. This blueprint may be applicable to PJI. The aim is to investigate Lactobacillus acidophilus spp. and their isolated extracellular-derived proteins (LaEPs) on PJI-relevant Staphylococcus aureus, methicillin-resistant S. aureus, and Escherichia coli planktonic growth and biofilm formation in vitro. The effect of LaEPs on cultured macrophages and osteogenic, and adipogenic human bone marrow-derived mesenchymal stem cell differentiation is analyzed. Data show electrostatically-induced probiotic-pathogen species co-aggregation and pathogenic growth inhibition together with LaEP-induced biofilm prevention. LaEPs prime macrophages for enhanced microbial phagocytosis via cathepsin K, reduce lipopolysaccharide-induced DNA damage and receptor activator nuclear factor-kappa B ligand expression, and promote a reparative M2 macrophage morphology under chronic inflammatory conditions. LaEPs also significantly augment bone deposition while abating adipogenesis thus holding promise as a potential multimodal therapeutic strategy. Proteomic analyses highlight high abundance of lysyl endopeptidase, and urocanate reductase. Further, in vivo analyses are warranted to elucidate their role in the prevention and treatment of PJIs.
Collapse
Affiliation(s)
| | - Anouska Seal
- Biionix Cluster, University of Central Florida, Orlando, FL, 32827, USA
| | | | - Udit Kumar
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), University of Central Florida, Orlando, FL, 32826, USA
| | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), University of Central Florida, Orlando, FL, 32826, USA
| | - Fei Wei
- Biionix Cluster, University of Central Florida, Orlando, FL, 32827, USA
| | | | - Melanie J Coathup
- Biionix Cluster, University of Central Florida, Orlando, FL, 32827, USA
- College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| |
Collapse
|
37
|
Faure-Dupuy S, Jubrail J, Depierre M, Africano-Gomez K, Öberg L, Israelsson E, Thörn K, Delevoye C, Castellano F, Herit F, Guilbert T, Russell DG, Mayer G, Cunoosamy DM, Kurian N, Niedergang F. ARL5b inhibits human rhinovirus 16 propagation and impairs macrophage-mediated bacterial clearance. EMBO Rep 2024; 25:1156-1175. [PMID: 38332148 PMCID: PMC10933434 DOI: 10.1038/s44319-024-00069-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: 05/01/2023] [Revised: 12/21/2023] [Accepted: 01/03/2024] [Indexed: 02/10/2024] Open
Abstract
Human rhinovirus is the most frequently isolated virus during severe exacerbations of chronic respiratory diseases, like chronic obstructive pulmonary disease. In this disease, alveolar macrophages display significantly diminished phagocytic functions that could be associated with bacterial superinfections. However, how human rhinovirus affects the functions of macrophages is largely unknown. Macrophages treated with HRV16 demonstrate deficient bacteria-killing activity, impaired phagolysosome biogenesis, and altered intracellular compartments. Using RNA sequencing, we identify the small GTPase ARL5b to be upregulated by the virus in primary human macrophages. Importantly, depletion of ARL5b rescues bacterial clearance and localization of endosomal markers in macrophages upon HRV16 exposure. In permissive cells, depletion of ARL5b increases the secretion of HRV16 virions. Thus, we identify ARL5b as a novel regulator of intracellular trafficking dynamics and phagolysosomal biogenesis in macrophages and as a restriction factor of HRV16 in permissive cells.
Collapse
Affiliation(s)
| | - Jamil Jubrail
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, 75014, France
- Southampton Solent University, East Park Terrace, Southampton, SO14 0YN, UK
| | - Manon Depierre
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, 75014, France
| | | | - Lisa Öberg
- Translational Science & Experimental Medicine, Research & Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 413 14, Sweden
| | - Elisabeth Israelsson
- Translational Science & Experimental Medicine, Research & Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 413 14, Sweden
| | - Kristofer Thörn
- Translational Science & Experimental Medicine, Research & Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 413 14, Sweden
| | - Cédric Delevoye
- Institut Curie, Université PSL, CNRS, UMR144, Structure and Membrane Compartments, Paris, France
- Institut Curie, Université PSL, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), Paris, France
| | - Flavia Castellano
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, 75014, France
- Université Paris Est Creteil, INSERM, IMRB, Creteil, 94010, France
| | - Floriane Herit
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, 75014, France
| | - Thomas Guilbert
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, 75014, France
| | - David G Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Gaell Mayer
- Immunology, Late stage Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 413 14, Sweden
| | - Danen M Cunoosamy
- Research & Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 413 14, Sweden
| | - Nisha Kurian
- Research & Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 413 14, Sweden
| | | |
Collapse
|
38
|
Bhatnagar A, Chopra U, Raja S, Das KD, Mahalingam S, Chakravortty D, Srinivasula SM. TLR-mediated aggresome-like induced structures comprise antimicrobial peptides and attenuate intracellular bacterial survival. Mol Biol Cell 2024; 35:ar34. [PMID: 38170582 PMCID: PMC10916861 DOI: 10.1091/mbc.e23-09-0347] [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: 09/05/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Immune cells employ diverse mechanisms for host defense. Macrophages, in response to TLR activation, assemble aggresome-like induced structures (ALIS). Our group has shown TLR4-signaling transcriptionally upregulates p62/sequestome1, which assembles ALIS. We have demonstrated that TLR4-mediated autophagy is, in fact, selective-autophagy of ALIS. We hypothesize that TLR-mediated autophagy and ALIS contribute to host-defense. Here we show that ALIS are assembled in macrophages upon exposure to different bacteria. These structures are associated with pathogen-containing phagosomes. Importantly, we present evidence of increased bacterial burden, where ALIS assembly is prevented with p62-specific siRNA. We have employed 3D-super-resolution structured illumination microscopy (3D-SR-SIM) and mass-spectrometric (MS) analyses to gain insight into the assembly of ALIS. Ultra-structural analyses of known constituents of ALIS (p62, ubiquitin, LC3) reveal that ALIS are organized structures with distinct patterns of alignment. Furthermore, MS-analyses of ALIS identified, among others, several proteins of known antimicrobial properties. We have validated MS data by testing the association of some of these molecules (Bst2, IFITM2, IFITM3) with ALIS and the phagocytosed-bacteria. We surmise that AMPs enrichment in ALIS leads to their delivery to bacteria-containing phagosomes and restricts the bacteria. Our findings in this paper support hitherto unknown functions of ALIS in host-defense.
Collapse
Affiliation(s)
- Anushree Bhatnagar
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India
| | - Umesh Chopra
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Sebastian Raja
- Laboratory of Molecular Cell Biology, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai 600036, India
| | - Krishanu Dey Das
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India
| | - S. Mahalingam
- Laboratory of Molecular Cell Biology, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai 600036, India
| | - Dipshikha Chakravortty
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Srinivasa Murty Srinivasula
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, Maruthamala PO, Vithura, Thiruvananthapuram 695551, Kerala, India
| |
Collapse
|
39
|
Hayashida N, Urano-Tashiro Y, Horie T, Saiki K, Yamanaka Y, Takahashi Y. Transcriptome and metabolome analyses of Streptococcus gordonii DL1 under acidic conditions. J Oral Biosci 2024; 66:112-118. [PMID: 38135272 DOI: 10.1016/j.job.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 12/24/2023]
Abstract
OBJECTIVES Streptococcus gordonii is associated with the formation of biofilms, especially those that comprise dental plaque. Notably, S. gordonii DL1 causes infective endocarditis (IE). Colonization of this bacterium requires a mechanism that can tolerate a drop in environmental pH by producing acid via its own sugar metabolism. The ability to survive acidic environmental conditions might allow the bacterium to establish vegetative colonization even in the endocardium due to inflammation-induced lowering of pH, increasing the risk of IE. At present, the mechanism by which S. gordonii DL1 survives under acidic conditions is not thoroughly elucidated. The present study was thus conducted to elucidate the mechanism(s) by which S. gordonii DL1 survives under acidic conditions. METHODS We analyzed dynamic changes in gene transcription and intracellular metabolites in S. gordonii DL1 exposed to acidic conditions, using transcriptome and metabolome analyses. RESULTS Transcriptome analysis revealed upregulation of genes involved in heat shock response and glycolysis, and down regulation of genes involved in phosphotransferase systems and biosynthesis of amino acids. The most upregulated genes were a beta-strand repeat protein of unknown function (SGO_RS06325), followed by copper-translocating P-type ATPase (SGO_RS09470) and malic enzyme (SGO_RS01850). The latter two of these contribute to cytoplasmic alkalinization. S. gordonii mutant strains lacking each of these genes showed significantly reduced survival under acidic conditions. Metabolome analysis revealed that cytoplasmic levels of several amino acids were reduced. CONCLUSIONS S. gordonii survives the acidic conditions by recovering the acidic cytoplasm using the various activities, which are regulated at the transcriptional level.
Collapse
Affiliation(s)
- Naoto Hayashida
- Department of Microbiology, The Nippon Dental University School of Life Dentistry at Tokyo, Japan.
| | - Yumiko Urano-Tashiro
- Department of Microbiology, The Nippon Dental University School of Life Dentistry at Tokyo, Japan.
| | - Tetsuro Horie
- Research Center for Odontology, The Nippon Dental University School of Life Dentistry at Tokyo, Japan.
| | - Keitarou Saiki
- Department of Microbiology, The Nippon Dental University School of Life Dentistry at Tokyo, Japan.
| | - Yuki Yamanaka
- Department of Microbiology, The Nippon Dental University School of Life Dentistry at Tokyo, Japan.
| | - Yukihiro Takahashi
- Department of Microbiology, The Nippon Dental University School of Life Dentistry at Tokyo, Japan.
| |
Collapse
|
40
|
Luo Y, Su L, Yang H, Geng A, Bai S, Zhou J. A disulfide molecule-vancomycin nanodrug delivery system efficiently eradicates intracellular bacteria. J Mater Chem B 2024; 12:2334-2345. [PMID: 38327236 DOI: 10.1039/d3tb02430j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Intracellular bacteria often lead to chronic and recurrent infections; however, most of the known antibiotics have poor efficacy against intracellular bacteria due to their poor cell membrane penetration efficiency into the cytosol. Here, a thiol-mediated nanodrug delivery system, named Van-DM NPs, was developed to improve vancomycin's penetration efficiency and intracellular antibacterial activities. Van-DM NPs were prepared through self-assembly of vancomycin (Van) and the disulfide molecule (DM) in NaOH buffer solution. On the one hand, the disulfide exchange reaction between Van-DM NPs and the bacterial surface enhances vancomycin accumulation in bacteria, increasing the local concentration of vancomycin. On the other hand, the disulfide exchange reaction between Van-DM NPs and the mammalian cell membrane triggered the translocation of Van-DM NPs across the mammalian cell membrane into the cell cytosol. These dual mechanisms promote antibacterial activities of vancomycin against both extracellular and intracellular bacteria S. aureus. Furthermore, in an intravenous S. aureus infection mouse model, Van-DM NPs exhibited high antibacterial capability and efficiently reduced the bacterial load in liver and spleen, where intracellular bacteria tend to reside. Altogether, the reported Van-DM NPs would be highly promising against intracellular pathogenic infections.
Collapse
Affiliation(s)
- Yuting Luo
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China.
| | - Liu Su
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China.
| | - Hui Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China.
| | - Aizhen Geng
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China.
| | - Shumeng Bai
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China.
| | - Jie Zhou
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China.
- China Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, The School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| |
Collapse
|
41
|
Ganio K, Nasreen M, Yang Z, Maunders EA, Luo Z, Hossain SI, Ngu DHY, Ellis D, Gu J, Neville SL, Wilksch J, Gunn AP, Whittall JJ, Kobe B, Deplazes E, Kappler U, McDevitt CA. Hfe Permease and Haemophilus influenzae Manganese Homeostasis. ACS Infect Dis 2024; 10:436-452. [PMID: 38240689 PMCID: PMC10863617 DOI: 10.1021/acsinfecdis.3c00407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 02/10/2024]
Abstract
Haemophilus influenzae is a commensal of the human upper respiratory tract that can infect diverse host niches due, at least in part, to its ability to withstand both endogenous and host-mediated oxidative stresses. Here, we show that hfeA, a gene previously linked to iron import, is essential for H. influenzae manganese recruitment via the HfeBCD transporter. Structural analyses show that metal binding in HfeA uses a unique mechanism that involves substantial rotation of the C-terminal lobe of the protein. Disruption of hfeA reduced H. influenzae manganese acquisition and was associated with decreased growth under aerobic conditions, impaired manganese-superoxide dismutase activity, reduced survival in macrophages, and changes in biofilm production in the presence of superoxide. Collectively, this work shows that HfeA contributes to H. influenzae manganese acquisition and virulence attributes. High conservation of the hfeABCD permease in Haemophilus species suggests that it may serve similar roles in other pathogenic Pasteurellaceae.
Collapse
Affiliation(s)
- Katherine Ganio
- Department
of Microbiology and Immunology, the Peter Doherty Institute for Infection
and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Marufa Nasreen
- School
of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
- Australian
Infectious Diseases Research Centre, The
University of Queensland, St Lucia, Queensland 4072, Australia
| | - Zihao Yang
- School
of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
- Australian
Infectious Diseases Research Centre, The
University of Queensland, St Lucia, Queensland 4072, Australia
| | - Eve A. Maunders
- Department
of Microbiology and Immunology, the Peter Doherty Institute for Infection
and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Zhenyao Luo
- School
of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
- Australian
Infectious Diseases Research Centre, The
University of Queensland, St Lucia, Queensland 4072, Australia
- Institute
for Molecular Bioscience, The University
of Queensland, St Lucia, Queensland 4072, Australia
| | - Sheikh Imamul Hossain
- School
of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
- School
of Life Sciences, University of Technology
Sydney, Ultimo, New South Wales 2007, Australia
| | - Dalton H. Y. Ngu
- School
of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
- Australian
Infectious Diseases Research Centre, The
University of Queensland, St Lucia, Queensland 4072, Australia
- Institute
for Molecular Bioscience, The University
of Queensland, St Lucia, Queensland 4072, Australia
| | - Daniel Ellis
- School
of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
- Australian
Infectious Diseases Research Centre, The
University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jin Gu
- School
of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
- Australian
Infectious Diseases Research Centre, The
University of Queensland, St Lucia, Queensland 4072, Australia
| | - Stephanie L. Neville
- Department
of Microbiology and Immunology, the Peter Doherty Institute for Infection
and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Jonathan Wilksch
- Department
of Microbiology and Immunology, the Peter Doherty Institute for Infection
and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Adam P. Gunn
- Department
of Microbiology and Immunology, the Peter Doherty Institute for Infection
and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Jonathan J. Whittall
- School of
Biological Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Boštjan Kobe
- School
of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
- Australian
Infectious Diseases Research Centre, The
University of Queensland, St Lucia, Queensland 4072, Australia
- Institute
for Molecular Bioscience, The University
of Queensland, St Lucia, Queensland 4072, Australia
| | - Evelyne Deplazes
- School
of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
- School
of Life Sciences, University of Technology
Sydney, Ultimo, New South Wales 2007, Australia
| | - Ulrike Kappler
- School
of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland 4072, Australia
- Australian
Infectious Diseases Research Centre, The
University of Queensland, St Lucia, Queensland 4072, Australia
| | - Christopher A. McDevitt
- Department
of Microbiology and Immunology, the Peter Doherty Institute for Infection
and Immunity, The University of Melbourne, Melbourne, Victoria 3000, Australia
| |
Collapse
|
42
|
Liu X, Yu B, Gao F, Jing P, Zhang P, Zheng G, Zhang X. Chemical immune conization of precancerous cervical lesions awakens immune cells and restores normal HPV negative and abnormal proliferation. Front Immunol 2024; 14:1259723. [PMID: 38235142 PMCID: PMC10791839 DOI: 10.3389/fimmu.2023.1259723] [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: 07/16/2023] [Accepted: 11/13/2023] [Indexed: 01/19/2024] Open
Abstract
Background Cervical cancer is one of the most common and deadly cancers in women, which is closely linked to the persistent infection of high-risk human papillomavirus (HPV). Current treatment of cervical cancer involves radical hysterectomy, radiotherapy, and chemotherapy or a combination. Objective We investigated if hapten-enhanced intratumoral chemotherapy (HEIC) was effective in boosting immunity for effective treatment of precancerous cervical lesions and HPV infection. Study design We used single-cell RNA sequencing (scRNA-Seq) to obtain transcriptome profiles of 40,239 cells from biopsies of precancerous cervical lesions from the cervix directly from one patient before the start of HEIC and approximately 1 week after HEIC. The blood samples were taken at the same time as biopsies. We compared the expression characteristics of malignant epithelial cells and immune cells, including epithelial cells, endothelial cells (ECs), fibroblasts, mural cells, T cells, B cells, T and NK neutrophils, mast cells, microparticles (MPs), and platelets, as well as the dynamic changes in cell percentage and cell subtype heterogeneity. Results Intratumoral injection of chemotherapy drug plus hapten induces an acute immune response in precancerous cervical lesions with HPV and further awakens immune cells to prevent the abnormal proliferation of the precancerous cells. Conclusion HEIC provides a potential treatment method for cervical cancer and HPV infection tailored to each patient's condition.
Collapse
Affiliation(s)
- Xueping Liu
- Department of Oncology, TaiMei Baofa Cancer Hospital, Dongping, Shandong, China
| | - Baofa Yu
- Department of Oncology, TaiMei Baofa Cancer Hospital, Dongping, Shandong, China
- Department of Oncology, Jinan Baofa Cancer Hospital, Jinan, Shandong, China
- Department of Oncology, Beijing Baofa Cancer Hospital, Beijing, China
- Department of Internal Medicine, South China Hospital of Shenzhen University, Shenzhen, China
- Core Lab., Immune Oncology Systems, Inc, San Diego, CA, United States
| | - Feng Gao
- Department of Oncology, TaiMei Baofa Cancer Hospital, Dongping, Shandong, China
| | - Peng Jing
- Department of Oncology, TaiMei Baofa Cancer Hospital, Dongping, Shandong, China
| | - Peicheng Zhang
- Department of Oncology, TaiMei Baofa Cancer Hospital, Dongping, Shandong, China
| | - Guoqin Zheng
- Department of Oncology, TaiMei Baofa Cancer Hospital, Dongping, Shandong, China
| | - Xiaomin Zhang
- Department of Oncology, TaiMei Baofa Cancer Hospital, Dongping, Shandong, China
| |
Collapse
|
43
|
Glorieux C, Enríquez C, González C, Aguirre-Martínez G, Buc Calderon P. The Multifaceted Roles of NRF2 in Cancer: Friend or Foe? Antioxidants (Basel) 2024; 13:70. [PMID: 38247494 PMCID: PMC10812565 DOI: 10.3390/antiox13010070] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/21/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Physiological concentrations of reactive oxygen species (ROS) play vital roles in various normal cellular processes, whereas excessive ROS generation is central to disease pathogenesis. The nuclear factor erythroid 2-related factor 2 (NRF2) is a critical transcription factor that regulates the cellular antioxidant systems in response to oxidative stress by governing the expression of genes encoding antioxidant enzymes that shield cells from diverse oxidative alterations. NRF2 and its negative regulator Kelch-like ECH-associated protein 1 (KEAP1) have been the focus of numerous investigations in elucidating whether NRF2 suppresses tumor promotion or conversely exerts pro-oncogenic effects. NRF2 has been found to participate in various pathological processes, including dysregulated cell proliferation, metabolic remodeling, and resistance to apoptosis. Herein, this review article will examine the intriguing role of phase separation in activating the NRF2 transcriptional activity and explore the NRF2 dual impacts on tumor immunology, cancer stem cells, metastasis, and long non-coding RNAs (LncRNAs). Taken together, this review aims to discuss the NRF2 multifaceted roles in both cancer prevention and promotion while also addressing the advantages, disadvantages, and limitations associated with modulating NRF2 therapeutically in cancer treatment.
Collapse
Affiliation(s)
- Christophe Glorieux
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Cinthya Enríquez
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique 1100000, Chile; (C.E.); (C.G.); (G.A.-M.)
- Programa de Magister en Ciencias Químicas y Farmacéuticas, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique 1100000, Chile
| | - Constanza González
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique 1100000, Chile; (C.E.); (C.G.); (G.A.-M.)
| | - Gabriela Aguirre-Martínez
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique 1100000, Chile; (C.E.); (C.G.); (G.A.-M.)
- Instituto de Química Medicinal, Universidad Arturo Prat, Iquique 1100000, Chile
| | - Pedro Buc Calderon
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique 1100000, Chile; (C.E.); (C.G.); (G.A.-M.)
- Instituto de Química Medicinal, Universidad Arturo Prat, Iquique 1100000, Chile
- Research Group in Metabolism and Nutrition, Louvain Drug Research Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| |
Collapse
|
44
|
Candeias E, Pereira-Santos AR, Empadinhas N, Cardoso SM, Esteves ARF. The Gut-Brain Axis in Alzheimer's and Parkinson's Diseases: The Catalytic Role of Mitochondria. J Alzheimers Dis 2024; 100:413-429. [PMID: 38875045 DOI: 10.3233/jad-240524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2024]
Abstract
Accumulating evidence suggests that gut inflammation is implicated in neuroinflammation in Alzheimer's and Parkinson's diseases. Despite the numerous connections it remains unclear how the gut and the brain communicate and whether gut dysbiosis is the cause or consequence of these pathologies. Importantly, several reports highlight the importance of mitochondria in the gut-brain axis, as well as in mechanisms like gut epithelium self-renewal, differentiation, and homeostasis. Herein we comprehensively address the important role of mitochondria as a cellular hub in infection and inflammation and as a link between inflammation and neurodegeneration in the gut-brain axis. The role of mitochondria in gut homeostasis and as well the crosstalk between mitochondria and gut microbiota is discussed. Significantly, we also review studies highlighting how gut microbiota can ultimately affect the central nervous system. Overall, this review summarizes novel findings regarding this cross-talk where the mitochondria has a main role in the pathophysiology of both Alzheimer's and Parkinson's disease strengthen by cellular, animal and clinical studies.
Collapse
Affiliation(s)
- Emanuel Candeias
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Ana Raquel Pereira-Santos
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Ph.D. Programme in Biomedicine and Experimental Biology (PDBEB), Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Nuno Empadinhas
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Sandra Morais Cardoso
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Raquel Fernandes Esteves
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
45
|
Sharma P, Venkatachalam K, Binesh A. Decades Long Involvement of THP-1 Cells as a Model for Macrophage Research: A Comprehensive Review. Antiinflamm Antiallergy Agents Med Chem 2024; 23:85-104. [PMID: 38676532 DOI: 10.2174/0118715230294413240415054610] [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: 01/03/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/29/2024]
Abstract
Over the years, researchers have endeavored to identify dependable and reproducible in vitro models for examining macrophage behavior under controlled conditions. The THP-1 cell line has become a significant and widely employed tool in macrophage research within these models. Originating from the peripheral blood of individuals with acute monocytic leukemia, this human monocytic cell line can undergo transformation into macrophage-like cells, closely mirroring primary human macrophages when exposed to stimulants. Macrophages play a vital role in the innate immune system, actively regulating inflammation, responding to infections, and maintaining tissue homeostasis. A comprehensive understanding of macrophage biology and function is crucial for gaining insights into immunological responses, tissue healing, and the pathogenesis of diseases such as viral infections, autoimmune disorders, and neoplastic conditions. This review aims to thoroughly evaluate and emphasize the extensive history of THP-1 cells as a model for macrophage research. Additionally, it will delve into the significance of THP-1 cells in advancing our comprehension of macrophage biology and their invaluable contributions to diverse scientific domains.
Collapse
Affiliation(s)
- Prakhar Sharma
- Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, 603103, Tamil Nadu, India
| | - Kaliyamurthi Venkatachalam
- Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, 603103, Tamil Nadu, India
| | - Ambika Binesh
- Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, 603103, Tamil Nadu, India
| |
Collapse
|
46
|
Case NT, Westman J, Hallett MT, Plumb J, Farheen A, Maxson ME, MacAlpine J, Liston SD, Hube B, Robbins N, Whitesell L, Grinstein S, Cowen LE. Respiration supports intraphagosomal filamentation and escape of Candida albicans from macrophages. mBio 2023; 14:e0274523. [PMID: 38038475 PMCID: PMC10746240 DOI: 10.1128/mbio.02745-23] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 10/16/2023] [Indexed: 12/02/2023] Open
Abstract
IMPORTANCE Candida albicans is a leading human fungal pathogen that often causes life-threatening infections in immunocompromised individuals. The ability of C. albicans to transition between yeast and filamentous forms is key to its virulence, and this occurs in response to many host-relevant cues, including engulfment by host macrophages. While previous efforts identified C. albicans genes required for filamentation in other conditions, the genes important for this morphological transition upon internalization by macrophages remained largely enigmatic. Here, we employed a functional genomic approach to identify genes that enable C. albicans filamentation within macrophages and uncovered a role for the mitochondrial ribosome, respiration, and the SNF1 AMP-activated kinase complex. Additionally, we showed that glucose uptake and glycolysis by macrophages support C. albicans filamentation. This work provides insights into the metabolic dueling that occurs during the interaction of C. albicans with macrophages and identifies vulnerabilities in C. albicans that could serve as promising therapeutic targets.
Collapse
Affiliation(s)
- Nicola T. Case
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Johannes Westman
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Jonathan Plumb
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Aiman Farheen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Michelle E. Maxson
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jessie MacAlpine
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Sean D. Liston
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoell Institute, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Sergio Grinstein
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Center of the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
47
|
Olivier FAB, Traven A. Quantitative live-cell imaging of Candida albicans escape from immune phagocytes. STAR Protoc 2023; 4:102737. [PMID: 37980567 PMCID: PMC10694764 DOI: 10.1016/j.xpro.2023.102737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/04/2023] [Accepted: 11/02/2023] [Indexed: 11/21/2023] Open
Abstract
Population-level dynamics of host-pathogen interactions can be characterized using quantitative live-cell imaging. Here, we present a protocol for infecting macrophages with the fungal pathogen Candida albicans in vitro and quantitative live-cell imaging of immune and pathogen responses. We describe steps for detailed image analysis and provide resources for quantification of phagocytosis and pathogen escape, as well as macrophage membrane permeabilization and viability. This protocol is modifiable for applications with a range of pathogens, immune cell types, and host-pathogen mechanisms. For complete details on the use and execution of this protocol, please refer to Olivier et al.1.
Collapse
Affiliation(s)
- Françios A B Olivier
- Infection Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia.
| | - Ana Traven
- Infection Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Centre to Impact AMR, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
48
|
Krampert L, Ossner T, Schröder A, Schatz V, Jantsch J. Simultaneous Increases in Intracellular Sodium and Tonicity Boost Antimicrobial Activity of Macrophages. Cells 2023; 12:2816. [PMID: 38132136 PMCID: PMC10741518 DOI: 10.3390/cells12242816] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
Inflamed and infected tissues can display increased local sodium (Na+) levels, which can have various effects on immune cells. In macrophages, high salt (HS) leads to a Na+/Ca2+-exchanger 1 (NCX1)-dependent increase in intracellular Na+ levels. This results in augmented osmoprotective signaling and enhanced proinflammatory activation, such as enhanced expression of type 2 nitric oxide synthase and antimicrobial function. In this study, the role of elevated intracellular Na+ levels in macrophages was investigated. Therefore, the Na+/K+-ATPase (NKA) was pharmacologically inhibited with two cardiac glycosides (CGs), ouabain (OUA) and digoxin (DIG), to raise intracellular Na+ without increasing extracellular Na+ levels. Exposure to HS conditions and treatment with both inhibitors resulted in intracellular Na+ accumulation and subsequent phosphorylation of p38/MAPK. The CGs had different effects on intracellular Ca2+ and K+ compared to HS stimulation. Moreover, the osmoprotective transcription factor nuclear factor of activated T cells 5 (NFAT5) was not upregulated on RNA and protein levels upon OUA and DIG treatment. Accordingly, OUA and DIG did not boost nitric oxide (NO) production and showed heterogeneous effects toward eliminating intracellular bacteria. While HS environments cause hypertonic stress and ionic perturbations, cardiac glycosides only induce the latter. Cotreatment of macrophages with OUA and non-ionic osmolyte mannitol (MAN) partially mimicked the HS-boosted antimicrobial macrophage activity. These findings suggest that intracellular Na+ accumulation and hypertonic stress are required but not sufficient to mimic boosted macrophage function induced by increased extracellular sodium availability.
Collapse
Affiliation(s)
- Luka Krampert
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg and University of Regensburg, 93053 Regensburg, Germany; (L.K.)
| | - Thomas Ossner
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg and University of Regensburg, 93053 Regensburg, Germany; (L.K.)
| | - Agnes Schröder
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg and University of Regensburg, 93053 Regensburg, Germany; (L.K.)
- Institute of Orthodontics, University Hospital Regensburg and University of Regensburg, 93053 Regensburg, Germany
| | - Valentin Schatz
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg and University of Regensburg, 93053 Regensburg, Germany; (L.K.)
| | - Jonathan Jantsch
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg and University of Regensburg, 93053 Regensburg, Germany; (L.K.)
- Institute for Medical Microbiology, Immunology, and Hygiene, Center for Molecular Medicine Cologne (CMMC), University Hospital Cologne and Faculty of Medicine, University of Cologne, 50935 Cologne, Germany
| |
Collapse
|
49
|
Jacobovitz MR, Hambleton EA, Guse A. Unlocking the Complex Cell Biology of Coral-Dinoflagellate Symbiosis: A Model Systems Approach. Annu Rev Genet 2023; 57:411-434. [PMID: 37722685 DOI: 10.1146/annurev-genet-072320-125436] [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] [Indexed: 09/20/2023]
Abstract
Symbiotic interactions occur in all domains of life, providing organisms with resources to adapt to new habitats. A prime example is the endosymbiosis between corals and photosynthetic dinoflagellates. Eukaryotic dinoflagellate symbionts reside inside coral cells and transfer essential nutrients to their hosts, driving the productivity of the most biodiverse marine ecosystem. Recent advances in molecular and genomic characterization have revealed symbiosis-specific genes and mechanisms shared among symbiotic cnidarians. In this review, we focus on the cellular and molecular processes that underpin the interaction between symbiont and host. We discuss symbiont acquisition via phagocytosis, modulation of host innate immunity, symbiont integration into host cell metabolism, and nutrient exchange as a fundamental aspect of stable symbiotic associations. We emphasize the importance of using model systems to dissect the cellular complexity of endosymbiosis, which ultimately serves as the basis for understanding its ecology and capacity to adapt in the face of climate change.
Collapse
Affiliation(s)
- Marie R Jacobovitz
- Cell Biology and Biophysics, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Elizabeth A Hambleton
- Division of Microbial Ecology, Department of Microbiology and Ecosystem Science, Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria;
| | - Annika Guse
- Faculty of Biology, Ludwig-Maximilians-Universität Munich, Munich, Germany;
| |
Collapse
|
50
|
Wang X, Sun B. Metabolic proteins with crucial roles in Edwardsiella tarda antioxidative adaptation and intracellular proliferation. mSystems 2023; 8:e0039123. [PMID: 37729581 PMCID: PMC10654080 DOI: 10.1128/msystems.00391-23] [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: 04/24/2023] [Accepted: 07/28/2023] [Indexed: 09/22/2023] Open
Abstract
IMPORTANCE Edwardsiella tarda is a significant fish pathogen that can live in challenging environments of reactive oxygen species (ROS), such as inside the phagocytes. Metabolic reconfiguration has been increasingly associated with bacterial oxidative tolerance and virulence. However, the metabolic proteins of E. tarda involved in such processes remain elusive. By proteomic analysis and functional characterization of protein null mutants, the present study identified eight crucial proteins for bacterial oxidative resistance and intracellular infection. Seven of them are metabolic proteins dictating the metabolic flux toward the generation of pyruvate, a key metabolite capable of scavenging ROS molecules. Furthermore, L-aspartate uptake, which can fuel the pyruvate generation, was found essential for the full antioxidative capacity of E. tarda. These findings identified seven metabolic proteins involved in bacterial oxidative adaptation and indicate that metabolic reprogramming toward pyruvate was likely a pivotal strategy of bacteria for antioxidative adaptation and intracellular survival.
Collapse
Affiliation(s)
- Xinhui Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
| | - Boguang Sun
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China
| |
Collapse
|