1
|
Li S, Huang Y, Sun Q, Li Y, Xie H, Fu Q. Caspase-1 is critical for mice in the defense against Streptococcus equi subsp. zooepidemicus infection by promoting macrophage phagocytosis. Microb Pathog 2025; 203:107499. [PMID: 40122410 DOI: 10.1016/j.micpath.2025.107499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Streptococcus equi subsp. zooepidemicus (SEZ) is an important pathogen which is responsible for a wide range of diseases in various species. Macrophages are professional phagocytes that can engulf microorganisms and trigger responses leading to microbial death. Caspase-1 is considered as a proinflammatory factor that mediates antibacterial response to protect hosts from bacteria. Here, we revealed a novel role of Caspase-1 in mice against SEZ. Through both in vitro and in vivo infection assays, we demonstrated that the maturation and secretion of the cytokine IL-1β are critically dependent on Caspase-1 activation. The Caspase-1 deficient mice displayed attenuation of bactericidal activity against SEZ, mainly by decreasing the accumulation of macrophage. In addition to the recruitment of macrophages, deficiency of Caspase-1 also impaired the phagocytosis of SEZ by macrophages. Our study demonstrated that Caspase-1 is critical for mice to defense against SEZ depending on the recruitment and phagocytosis of macrophage.
Collapse
Affiliation(s)
- Shun Li
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Yunfei Huang
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Qinqin Sun
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Yajuan Li
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Honglin Xie
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China.
| | - Qiang Fu
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China.
| |
Collapse
|
2
|
Fei X, Li N, Xu X, Zhu Y. Macrophage biology in the pathogenesis of Helicobacter pylori infection. Crit Rev Microbiol 2025; 51:399-416. [PMID: 39086061 DOI: 10.1080/1040841x.2024.2366944] [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: 08/03/2023] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 08/02/2024]
Abstract
Infection with H. pylori induces chronic gastric inflammation, progressing to peptic ulcer and stomach adenocarcinoma. Macrophages function as innate immune cells and play a vital role in host immune defense against bacterial infection. However, the distinctive mechanism by which H. pylori evades phagocytosis allows it to colonize the stomach and further aggravate gastric preneoplastic pathology. H. pylori exacerbates gastric inflammation by promoting oxidative stress, resisting macrophage phagocytosis, and inducing M1 macrophage polarization. M2 macrophages facilitate the proliferation, invasion, and migration of gastric cancer cells. Various molecular mechanisms governing macrophage function in the pathogenesis of H. pylori infection have been identified. In this review, we summarize recent findings of macrophage interactions with H. pylori infection, with an emphasis on the regulatory mechanisms that determine the clinical outcome of bacterial infection.
Collapse
Affiliation(s)
- Xiao Fei
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Nianshuang Li
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xinbo Xu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yin Zhu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Digestive Diseases, Department of Gastroenterology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
3
|
Heroor A, Dandekar P, Murthy SI. Outcomes of a Tenon Patch Graft for Corneal Perforations in Resolving Infective Keratitis. Cornea 2025:00003226-990000000-00816. [PMID: 39937047 DOI: 10.1097/ico.0000000000003834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/01/2025] [Indexed: 02/13/2025]
Abstract
PURPOSE To report the outcomes of a Tenon patch graft (TPG) in sealing corneal perforations in cases with resolving infective keratitis. METHODS This retrospective interventional study was conducted at a tertiary eye care center. All patients who underwent a TPG for corneal perforations because of clinically or/and microbiologically diagnosed infective keratitis between 2021 and 2023 were included. The surgical technique was consistent with that used in noninfective etiology. Out of 100 cases undergoing a TPG during the same period, 15 eyes of 14 patients had corneal perforations secondary to infective keratitis. RESULTS The etiological profile included bacterial keratitis in 8, viral and fungal keratitis in 3, and Pythium keratitis in 1 eye of 15 eyes. One patient had bilateral infective keratitis after refractive surgery. The perforations were central in 7 and paracentral in 8 eyes. Successful restoration of tectonic integrity at 1 month was achieved in 14/15 eyes (93%). One patient had a repeat perforation at 3 weeks postoperatively, which was managed with a cyanoacrylate glue application. Infection control was achieved in all cases with adjunctive topical antimicrobial therapy. CONCLUSIONS A TPG is a viable option for restoring globe integrity in corneal perforations encountered in resolving infective keratitis.
Collapse
Affiliation(s)
- Aniruddh Heroor
- Academy of Eye Care, LV Prasad Eye Institute, Hyderabad, India
| | - Prajakta Dandekar
- Cornea and Anterior Segment Services, Shantilal Shanghvi Eye Institute, Mumbai, India; and
| | - Somasheila I Murthy
- Cornea and Anterior Segment Services, Shantilal Shanghvi Eye Institute, Mumbai, India; and
- Shantilal Shanghvi Cornea Institute, LV Prasad Eye Institute, Hyderabad, India
| |
Collapse
|
4
|
Huang W, Yuan H, Yang H, Shen Y, Guo L, Zhong N, Wu T, Shen Y, Chen G, Huang S, Niu L, Ouyang G. A Hierarchical Metal-Organic Framework Intensifying ROS Catalytic Activity and Bacterial Entrapment for Engineering Self-Antimicrobial Mask. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410703. [PMID: 39686695 PMCID: PMC11809350 DOI: 10.1002/advs.202410703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/14/2024] [Indexed: 12/18/2024]
Abstract
Leveraging functional materials to develop advanced personal protective equipment is of significant importance for cutting off the propagation of infectious diseases, yet faces ongoing challenges owing to the unsatisfied antimicrobial efficiency. Herein a hierarchically porous cerium metal-organic framework (Ce-MOF) boosting the antimicrobial performance by intensifying catalytic reactive oxygen species (ROS) generation and bacterial entrapment simultaneously is reported. This Ce-MOF presents dendritic surface topography and hierarchical pore channels where the Lewis acid Ce sites are dispersedly anchored. Attributing to this sophisticated nanoarchitecture rendering the catalytic Ce sites highly accessible, it shows a ca. 1800-fold activity enhancement for the catalytic conversion of atmospheric oxygen to highly toxic ROS compared to traditional CeO2. Additionally, the dendritic and negative-charged surface engineered in this Ce-MOF substantially enhances the binding affinity toward positive-charged bacteria, enabling the spatial proximity between the bacteria and the short-lived ROS and therefore maximizing the utilization of highly toxic ROS to inactivate bacteria. It is demonstrated that this Ce-MOF-integrated face mask displays almost 100% antimicrobial efficacy even in insufficient light and dark scenarios. This work provides important insights into the design of antibacterial MOF materials by a pore- and surface-engineering strategy and sheds new light on the development of advanced self-antimicrobial devices.
Collapse
Affiliation(s)
- Wei Huang
- School of Chemical Engineering and TechnologySouthern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Sun Yat‐sen UniversityZhuhai519082P. R. China
| | - Haitao Yuan
- Center for Drug Research and Development Guangdong Provincial Key Laboratory of Advanced Drug Delivery SystemGuangdong Pharmaceutical UniversityGuangzhou510006P. R. China
| | - Huangsheng Yang
- School of ChemistrySun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Yujian Shen
- School of ChemistrySun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Lihong Guo
- School of ChemistrySun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Ningyi Zhong
- School of ChemistrySun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Tong Wu
- Department of RadiologyThe Third Affiliated Hospital of Southern Medical UniversitySouthern Medical UniversityGuangzhou510630P. R. China
| | - Yong Shen
- School of ChemistrySun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Guosheng Chen
- School of Chemical Engineering and TechnologySouthern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Sun Yat‐sen UniversityZhuhai519082P. R. China
- School of ChemistrySun Yat‐sen UniversityGuangzhou510006P. R. China
- Sun Yat‐sen University School of Chemistry and Guangdong Basic Research Center of Excellence for Functional Molecular EngineeringSun Yat‐sen UniversityGuangzhou510006P. R. China
| | - Siming Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacologythe NMPA and State Key Laboratory of Respiratory DiseaseSchool of Pharmaceutical Sciences and the Fifth Affiliated HospitalGuangzhou Medical UniversityGuangzhou511436P. R. China
| | - Li Niu
- School of Chemical Engineering and TechnologySouthern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Sun Yat‐sen UniversityZhuhai519082P. R. China
| | - Gangfeng Ouyang
- School of Chemical Engineering and TechnologySouthern Marine Science and Engineering Guangdong Laboratory (Zhuhai)Sun Yat‐sen UniversityZhuhai519082P. R. China
- School of ChemistrySun Yat‐sen UniversityGuangzhou510006P. R. China
- Sun Yat‐sen University School of Chemistry and Guangdong Basic Research Center of Excellence for Functional Molecular EngineeringSun Yat‐sen UniversityGuangzhou510006P. R. China
| |
Collapse
|
5
|
Chen X, Shi X, Liu X, Zhai X, Li W, Hong W. Eliminating Intracellular MRSA via Mannosylated Lipid-Coated Calcium Phosphate Nanoparticles. Mol Pharm 2024; 21:5772-5783. [PMID: 39368111 DOI: 10.1021/acs.molpharmaceut.4c00779] [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: 10/07/2024]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) within cells proves exceptionally challenging to eradicate using conventional antimicrobials, resulting in recurring infections and heightened resistance. Herein, we reported an innovative mannosylated lipid-coated photodynamic/photothermal calcium phosphate nanoparticle (MAN-LCaP@ICG) for eradicating intracellular MRSA. The MAN-LCaP functioned as the vehicle for drug delivery, exhibiting preferential uptake by macrophages and facilitating the transport of ICG to intracellular pathogens. The MAN units integrated into MAN-LCaP@ICG could promote binding with MAN residuals on macrophage cells, as evidenced by cellular uptake assays using fluorescence microscopy and flow cytometry. Following its targeted accumulation, MAN-LCaP@ICG could enter into the cytoplasm and efficiently eradicate intracellular MRSA by a combination of the lysosome escape capability of CaP and the photodynamic and photothermal therapeutic effects of ICG. Furthermore, MAN-LCaP@ICG could kill MRSA more effectively than LCaP@ICG without MAN units or free ICG in a mouse peritoneal infection model. Therefore, MAN-LCaP@ICG provided a promising direction for human clinical application in combating intracellular infections.
Collapse
Affiliation(s)
- Xiangjun Chen
- School of Pharmacy, Shandong Engineering Research Center of New-Type Drug Loading & Releasing Technology and Preparation, Binzhou Medical University, 346 Guanhai Road, Yantai 264003, P. R. China
| | - Xiaoyi Shi
- School of Pharmacy, Shandong Engineering Research Center of New-Type Drug Loading & Releasing Technology and Preparation, Binzhou Medical University, 346 Guanhai Road, Yantai 264003, P. R. China
| | - Xiao Liu
- School of Pharmacy, Shandong Engineering Research Center of New-Type Drug Loading & Releasing Technology and Preparation, Binzhou Medical University, 346 Guanhai Road, Yantai 264003, P. R. China
| | - Xuanxiang Zhai
- School of Pharmacy, Shandong Engineering Research Center of New-Type Drug Loading & Releasing Technology and Preparation, Binzhou Medical University, 346 Guanhai Road, Yantai 264003, P. R. China
| | - Wenting Li
- School of Pharmacy, Shandong Engineering Research Center of New-Type Drug Loading & Releasing Technology and Preparation, Binzhou Medical University, 346 Guanhai Road, Yantai 264003, P. R. China
| | - Wei Hong
- School of Pharmacy, Shandong Engineering Research Center of New-Type Drug Loading & Releasing Technology and Preparation, Binzhou Medical University, 346 Guanhai Road, Yantai 264003, P. R. China
| |
Collapse
|
6
|
Hu M, Jin F, Zhang C, Shao J, Wang C, Wang T, Wu D. Sodium houttuyfonate induces bacterial lipopolysaccharide shedding to promote macrophage M1 polarization against acute bacterial lung infection. Biomed Pharmacother 2024; 179:117358. [PMID: 39278188 DOI: 10.1016/j.biopha.2024.117358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/18/2024] Open
Abstract
Sodium houttuyfonate (SH), derived from the widely utilized natural herb Houttuynia cordata, exhibits an effective therapeutic effect on various diseases, including bacterial and fungal infections, especially the respiratory tract infection. Therefore, the anti-microbial mechanisms of SH may be different from the single-target action mechanism of conventional antibiotics, and further research is needed to clarify this. Firstly, we discovered that SH can effectively intervene in mouse lung infections by reducing bacterial load and acute inflammation response related to pneumonia caused by Pseudomonas aeruginosa. Interestingly, our results confirmed that SH has surface activity and can directly induce changes in the cell wall the shedding of surface lipopolysaccharide (LPS). Additionally, we found that SH-induced shedding of LPS can induce M1 polarization of macrophages in the early stage, leading to the production of corresponding polarization effector molecules. Subsequently, we discovered that SH-induced M1 polarization cells can effectively phagocytose and kill bacterial cells. The protein expression results indicated that SH can enhance the expression of M1 polarization pathway of TLR4/MyD88/NF-κB during the initial phase of macrophage and pathogen interaction. In summary, our results imply that SH could directly induce the shedding of P. aeruginosa LPS in a surfactant-like manner. Afterwards, the SH induced abscisic LPS can initiate the TLR4/MyD88/NF-κB immune pathway to trigger the M1 polarization of macrophages, which might intervene the P. aeruginosa-caused acute lung infection at early stage. Based on these findings, we attempted to coin the term "immune feedback eradication mechanism against pathogen of natural product" to describe this potent antimicrobial mechanism of SH.
Collapse
Affiliation(s)
- Mengxue Hu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, China
| | - Feng Jin
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, China
| | - Cangcang Zhang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, China
| | - Jing Shao
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, 350 Longzihu Road, Hefei 230012, China
| | - Changzhong Wang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, 350 Longzihu Road, Hefei 230012, China
| | - Tianming Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, 350 Longzihu Road, Hefei 230012, China.
| | - Daqiang Wu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Hefei 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, 350 Longzihu Road, Hefei 230012, China.
| |
Collapse
|
7
|
Cerdeira CD, Brigagão MRPL. Targeting Macrophage Polarization in Infectious Diseases: M1/M2 Functional Profiles, Immune Signaling and Microbial Virulence Factors. Immunol Invest 2024; 53:1030-1091. [PMID: 38913937 DOI: 10.1080/08820139.2024.2367682] [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: 06/26/2024]
Abstract
INTRODUCTION An event of increasing interest during host-pathogen interactions is the polarization of patrolling/naive monocytes (MOs) into macrophage subsets (MФs). Therapeutic strategies aimed at modulating this event are under investigation. METHODS This review focuses on the mechanisms of induction/development and profile of MФs polarized toward classically proinflammatory (M1) or alternatively anti-inflammatory (M2) phenotypes in response to bacteria, fungi, parasites, and viruses. RESULTS AND DISCUSSION It highlights nuclear, cytoplasmic, and cell surface receptors (pattern recognition receptors/PPRs), microenvironmental mediators, and immune signaling. MФs polarize into phenotypes: M1 MФs, activated by IFN-γ, pathogen-associated molecular patterns (PAMPs, e.g. lipopolysaccharide) and membrane-bound PPRs ligands (TLRs/CLRs ligands); or M2 MФs, induced by interleukins (ILs-4, -10 and -13), antigen-antibody complexes, and helminth PAMPs. Polarization toward M1 and M2 profiles evolve in a pathogen-specific manner, with or without canonicity, and can vary widely. Ultimately, this can result in varying degrees of host protection or more severe disease outcome. On the one hand, the host is driving effective MФs polarization (M1 or M2); but on the other hand, microorganisms may skew the polarization through virulence factors to increase pathogenicity. Cellular/genomic reprogramming also ensures plasticity of M1/M2 phenotypes. Because modulation of polarization can occur at multiple points, new insights and emerging perspectives may have clinical implications during the inflammation-to-resolution transition; translated into practical applications as for therapeutic/vaccine design target to boost microbicidal response (M1, e.g. triggering oxidative burst) with specifics PAMPs/IFN-γ or promote tissue repair (M2, increasing arginase activity) via immunotherapy.
Collapse
|
8
|
Li X, Chen RY, Shi JJ, Li CY, Liu YJ, Gao C, Gao MR, Zhang S, Lu JF, Cao JF, Yang GJ, Chen J. Emerging role of Jumonji domain-containing protein D3 in inflammatory diseases. J Pharm Anal 2024; 14:100978. [PMID: 39315124 PMCID: PMC11417268 DOI: 10.1016/j.jpha.2024.100978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 09/25/2024] Open
Abstract
Jumonji domain-containing protein D3 (JMJD3) is a 2-oxoglutarate-dependent dioxygenase that specifically removes transcriptional repression marks di- and tri-methylated groups from lysine 27 on histone 3 (H3K27me2/3). The erasure of these marks leads to the activation of some associated genes, thereby influencing various biological processes, such as development, differentiation, and immune response. However, comprehensive descriptions regarding the relationship between JMJD3 and inflammation are lacking. Here, we provide a comprehensive overview of JMJD3, including its structure, functions, and involvement in inflammatory pathways. In addition, we summarize the evidence supporting JMJD3's role in several inflammatory diseases, as well as the potential therapeutic applications of JMJD3 inhibitors. Additionally, we also discuss the challenges and opportunities associated with investigating the functions of JMJD3 and developing targeted inhibitors and propose feasible solutions to provide valuable insights into the functional exploration and discovery of potential drugs targeting JMJD3 for inflammatory diseases.
Collapse
Affiliation(s)
- Xiang Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang, 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Ru-Yi Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang, 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Jin-Jin Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang, 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Chang-Yun Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang, 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Yan-Jun Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang, 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Chang Gao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang, 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Ming-Rong Gao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang, 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Shun Zhang
- Ningbo No. 2 Hospital, Ningbo, Zhejiang, 315211, China
- China Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, 315211, China
| | - Jian-Fei Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang, 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Jia-Feng Cao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang, 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang, 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, China
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang, 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315211, China
| |
Collapse
|
9
|
Luo L, Wang H, Xiong J, Chen X, Shen X, Zhang H. Echinatin attenuates acute lung injury and inflammatory responses via TAK1-MAPK/NF-κB and Keap1-Nrf2-HO-1 signaling pathways in macrophages. PLoS One 2024; 19:e0303556. [PMID: 38753858 PMCID: PMC11098428 DOI: 10.1371/journal.pone.0303556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/27/2024] [Indexed: 05/18/2024] Open
Abstract
Echinatin is an active ingredient in licorice, a traditional Chinese medicine used in the treatment of inflammatory disorders. However, the protective effect and underlying mechanism of echinatin against acute lung injury (ALI) is still unclear. Herein, we aimed to explore echinatin-mediated anti-inflammatory effects on lipopolysaccharide (LPS)-stimulated ALI and its molecular mechanisms in macrophages. In vitro, echinatin markedly decreased the levels of nitric oxide (NO) and prostaglandin E2 (PGE2) in LPS-stimulated murine MH-S alveolar macrophages and RAW264.7 macrophages by suppressing inducible nitric oxide synthase and cyclooxygenase-2 (COX-2) expression. Furthermore, echinatin reduced LPS-induced mRNA expression and release of interleukin-1β (IL-1β) and IL-6 in RAW264.7 cells. Western blotting and CETSA showed that echinatin repressed LPS-induced activation of mitogen-activated protein kinase (MAPK) and nuclear factor-kappa B (NF-κB) pathways through targeting transforming growth factor-beta-activated kinase 1 (TAK1). Furthermore, echinatin directly interacted with Kelch-like ECH-associated protein 1 (Keap1) and activated the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway to enhance heme oxygenase-1 (HO-1) expression. In vivo, echinatin ameliorated LPS-induced lung inflammatory injury, and reduced production of IL-1β and IL-6. These findings demonstrated that echinatin exerted anti-inflammatory effects in vitro and in vivo, via blocking the TAK1-MAPK/NF-κB pathway and activating the Keap1-Nrf2-HO-1 pathway.
Collapse
Affiliation(s)
- Liuling Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinrui Xiong
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaorui Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofei Shen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hai Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Bhardwaj C, Srivastava P. Identification of hub genes in placental dysfunction and recurrent pregnancy loss through transcriptome data mining: A meta-analysis. Taiwan J Obstet Gynecol 2024; 63:297-306. [PMID: 38802191 DOI: 10.1016/j.tjog.2024.01.035] [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] [Accepted: 01/30/2024] [Indexed: 05/29/2024] Open
Abstract
Recurrent pregnancy loss (RPL) is a condition characterized by the loss of two or more pregnancies before 20 weeks of gestation. The causes of RPL are complex and can be due to a variety of factors, including genetic, immunological, hormonal, and environmental factors. This transcriptome data mining study was done to explore the differentially expressed genes (DEGs) and related pathways responsible for pathogenesis of RPL using an Insilco approach. RNAseq datasets from the Gene Expression Omnibus (GEO) database was used to extract RNAseq datasets of RPL. Meta-analysis was done by ExpressAnalyst. The functional and pathway enrichment analysis of DEGs were performed using KEGG and BINGO plugin of Cytoscape software. Protein-protein interaction was done using STRING and hub genes were identified. A total of 91 DEGs were identified, out of which 10 were downregulated and 81 were upregulated. Pathway analysis indicated that majority of DEGs were enriched in immunological pathways (IL-17 signalling pathway, TLR-signalling pathway, autoimmune thyroid disease), angiogenic VEGF-signalling pathway and cell-cycle signalling pathways. Of these, 10 hub genes with high connectivity were selected (CXCL8, CCND1, FOS, PTGS2, CTLA4, THBS1, MMP2, KDR, and CD80). Most of these genes are involved in maintenance of immune response at maternal-fetal interface. Further, in functional enrichment analyses revealed the highest node size in regulation of biological processes followed by cellular processes, their regulation and regulation of multicellular organismal process. This in-silico transcriptomics meta-analysis findings could potentially contribute in identifying novel biomarkers and therapeutic targets for RPL, which could lead to the development of new diagnostic and therapeutic strategies for this condition.
Collapse
Affiliation(s)
- Chitra Bhardwaj
- Genetic Metabolic Unit, Department of Paediatrics, Advanced Paediatrics Centre, Post Graduate Institute of Medical Education & Research, Sector-12, Chandigarh, 160012, India
| | - Priyanka Srivastava
- Genetic Metabolic Unit, Department of Paediatrics, Advanced Paediatrics Centre, Post Graduate Institute of Medical Education & Research, Sector-12, Chandigarh, 160012, India.
| |
Collapse
|
11
|
Wei YF, Xie SA, Zhang ST. Current research on the interaction between Helicobacter pylori and macrophages. Mol Biol Rep 2024; 51:497. [PMID: 38598010 DOI: 10.1007/s11033-024-09395-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/28/2024] [Indexed: 04/11/2024]
Abstract
Helicobacter pylori (H. pylori) is a gram-negative bacteria with a worldwide infection rate of 50%, known to induce gastritis, ulcers and gastric cancer. The interplay between H. pylori and immune cells within the gastric mucosa is pivotal in the pathogenesis of H. pylori-related disease. Following H. pylori infection, there is an observed increase in gastric mucosal macrophages, which are associated with the progression of gastritis. H. pylori elicits macrophage polarization, releases cytokines, reactive oxygen species (ROS) and nitric oxide (NO) to promote inflammatory response and eliminate H. pylori. Meanwhile, H. pylori has developed mechanisms to evade the host immune response in order to maintain the persistent infection, including interference with macrophage phagocytosis and antigen presentation, as well as induction of macrophage apoptosis. Consequently, the interaction between H. pylori and macrophages can significantly impact the progression, pathogenesis, and resolution of H. pylori infection. Moreover, macrophages are emerging as potential therapeutic targets for H. pylori-associated gastritis. Therefore, elucidating the involvement of macrophages in H. pylori infection may provide novel insights into the pathogenesis, progression, and management of H. pylori-related disease.
Collapse
Affiliation(s)
- Yan-Fei Wei
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Si-An Xie
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
- Beijing Digestive Disease Center, State Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Disease, Beijing, 100050, China.
| | - Shu-Tian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
12
|
Wang Z, Liu T, Wang Z, Mi Z, Zhang Y, Wang C, Sun L, Ma S, Xue X, Liu H, Zhang F. CYBB-Mediated Ferroptosis Associated with Immunosuppression in Mycobacterium leprae-Infected Monocyte-Derived Macrophages. J Invest Dermatol 2024; 144:874-887.e2. [PMID: 37925067 DOI: 10.1016/j.jid.2023.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 11/06/2023]
Abstract
Mycobacterium leprae-infected macrophages preferentially exhibit the regulatory M2 phenotype in vitro, which helps the immune escape unabated growth of M leprae in host cells. The mechanism that triggers macrophage polarization is still unknown. In this study, we performed single-cell RNA sequencing to determine the initial responses of human monocyte-derived macrophages against M leprae infection of 4 healthy individuals and found an increase in a major alternative-activated macrophage type that overexpressed NEAT1, CCL2, and CD163. Importantly, further functional analysis showed that ferroptosis was positively correlated with M2 polarization of macrophages, and in vitro experiments have shown that inhibition of ferroptosis promotes the survival of M leprae within macrophages. In addition, further joint analysis of our results with mutisequencing data from patients with leprosy and in vitro validation identified that CYBB was the pivotal molecule for ferroptosis that could promote the M2 polarization of M leprae-infected macrophages, resulting in the immune escape and unabated growth of pathogenic bacteria. Overall, our results suggest that M leprae facilitated its survival by inducing CYBB-mediated macrophage ferroptosis leading to its alternative activation and might reveal the potential for a new therapeutic strategy of leprosy.
Collapse
Affiliation(s)
- Zhe Wang
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Tingting Liu
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Zhenzhen Wang
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Zihao Mi
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yuan Zhang
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Chuan Wang
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Lele Sun
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Shanshan Ma
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaotong Xue
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Hong Liu
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
| | - Furen Zhang
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Venereology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
13
|
Zhu Y, Jin X, Fu N, Li J. Medrysone promotes corneal injury repair by promoting M2-like polarization of macrophages. BMC Ophthalmol 2023; 23:503. [PMID: 38082280 PMCID: PMC10712160 DOI: 10.1186/s12886-023-03234-3] [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: 06/11/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Accumulated evidence suggests that M2-like polarized macrophages plays an important role in reducing inflammation, promoting and accelerating wound healing process and tissue repair. Thus, M2-like TAMs (Tumour-associated macrophages) was an appealing target for therapy intervention. METHOD Flow cytometry and RT-PCR assay were used to detect the polarization of macrophages induced by Medrysone, and the rat corneal mechanical injury model was established to evaluate the efficacy of Medrysone in cornel repair. RESULTS Here we found that Medrysone enhanced IL-4 induced M2 polarization of macrophages, as illustrated by increased expression of CD206, up-regulation of M2 marker mRNAs. Medrysone promoted VEGF and CCL2 secretion in IL-4 induced M2-like polarization. IL-4 triggered STAT6 activation was further enhanced by Medrysone and silencing of STAT6 partially abrogated the stimulatory effect of Medrysone. Medrysone improved migration-promoting feature of M2-like macrophages, as indicated by increased migration of endothelial cells. Further, Medrysone promoted corneal injury repair by inducing M2 polarization of macrophages in vivo. CONCLUSION Our study suggest that Medrysone promotes corneal injury repair by inducing the M2 polarization of macrophages, providing a theoretical basis for the application of Medrysone in the treatment of corneal injury.
Collapse
Affiliation(s)
- Yaqin Zhu
- Hangzhou Aier Eye Hospital, Hangzhou, 310000, Zhejiang, China
| | - Xiaohong Jin
- Hangzhou Aier Eye Hospital, Hangzhou, 310000, Zhejiang, China
| | - Ning Fu
- Sir Run Run Shaw Hospital Hangzhou, Hangzhou, 310000, China
| | - Jiuke Li
- Hangzhou Aier Eye Hospital, Hangzhou, 310000, Zhejiang, China.
| |
Collapse
|
14
|
Heo J, Park YJ, Kim Y, Lee HS, Kim J, Kwon SH, Kang MG, Rhee HW, Sun W, Lee JH, Cho H. Mitochondrial E3 ligase MARCH5 is a safeguard against DNA-PKcs-mediated immune signaling in mitochondria-damaged cells. Cell Death Dis 2023; 14:788. [PMID: 38040710 PMCID: PMC10692114 DOI: 10.1038/s41419-023-06315-9] [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: 04/04/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023]
Abstract
Mitochondrial dysfunction is important in various chronic degenerative disorders, and aberrant immune responses elicited by cytoplasmic mitochondrial DNA (mtDNA) may be related. Here, we developed mtDNA-targeted MTERF1-FokI and TFAM-FokI endonuclease systems to induce mitochondrial DNA double-strand breaks (mtDSBs). In these cells, the mtDNA copy number was significantly reduced upon mtDSB induction. Interestingly, in cGAS knockout cells, synthesis of interferon β1 and interferon-stimulated gene was increased upon mtDSB induction. We found that mtDSBs activated DNA-PKcs and HSPA8 in a VDAC1-dependent manner. Importantly, the mitochondrial E3 ligase MARCH5 bound active DNA-PKcs in cells with mtDSBs and reduced the type І interferon response through the degradation of DNA-PKcs. Likewise, mitochondrial damage caused by LPS treatment in RAW264.7 macrophage cells increased phospho-HSPA8 levels and the synthesis of mIFNB1 mRNA in a DNA-PKcs-dependent manner. Accordingly, in March5 knockout macrophages, phospho-HSPA8 levels and the synthesis of mIFNB1 mRNA were prolonged after LPS stimulation. Together, cytoplasmic mtDNA elicits a cellular immune response through DNA-PKcs, and mitochondrial MARCH5 may be a safeguard to prevent persistent inflammatory reactions.
Collapse
Affiliation(s)
- June Heo
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
| | - Yeon-Ji Park
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, South Korea
| | - Yonghyeon Kim
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
| | - Ho-Soo Lee
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, South Korea
| | - Jeongah Kim
- Department of Anatomy, College of medicine, Korea University, Seoul, South Korea
| | - Soon-Hwan Kwon
- Department of Infectious Diseases, Research Center of Infectious and Environmental Diseases, Armed Forces Medical Research Institute, Daejeon, South Korea
| | - Myeong-Gyun Kang
- Department of Chemistry, Seoul National University, Seoul, South Korea
| | - Hyun-Woo Rhee
- Department of Chemistry, Seoul National University, Seoul, South Korea
| | - Woong Sun
- Department of Anatomy, College of medicine, Korea University, Seoul, South Korea
| | - Jae-Ho Lee
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, South Korea.
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea.
| | - Hyeseong Cho
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, South Korea.
| |
Collapse
|
15
|
Song X, Duan R, Duan L, Wei L. Current knowledge of the immune reconstitution inflammatory syndrome in Whipple disease: a review. Front Immunol 2023; 14:1265414. [PMID: 37901208 PMCID: PMC10611461 DOI: 10.3389/fimmu.2023.1265414] [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: 07/22/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
Immune reconstitution inflammatory syndrome (IRIS) is characterized by exaggerated and dysregulated inflammatory responses that occur as a result of reconstitution of adaptive or innate immunity. A wide range of microorganisms have been found to be associated with IRIS, such as human immunodeficiency virus (HIV), Mycobacterium and actinobacteria. Whipple disease (WD) is an infectious disorder caused by the Gram-positive bacterium Tropheryma whipplei (T. whipplei) and IRIS also serves as a complication during its treament. Although many of these pathological mechanisms are shared with related inflammatory disorders, IRIS in WD exhibits distinct features and is poorly described in the medical literature. Novel investigations of the intestinal mucosal immune system have provided new insights into the pathogenesis of IRIS, elucidating the interplay between systemic and local immune responses. These insights may be used to identify monitoring tools for disease prevention and to develop treatment strategies. Therefore, this review synthesizes these new concepts in WD IRIS to approach the feasibility of manipulating host immunity and immune reconstitution of inflammatory syndromes from a newer, more comprehensive perspective and study hypothetical options for the management of WD IRIS.
Collapse
Affiliation(s)
| | | | | | - Lijuan Wei
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Chang Chun, Jilin, China
| |
Collapse
|
16
|
Quaresma TC, de Aguiar Valentim L, de Sousa JR, de Souza Aarão TL, Fuzii HT, Duarte MIS, de Souza J, Quaresma JAS. Immunohistochemical Characterization of M1, M2, and M4 Macrophages in Leprosy Skin Lesions. Pathogens 2023; 12:1225. [PMID: 37887741 PMCID: PMC10610015 DOI: 10.3390/pathogens12101225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Mycobacterium leprae is the etiological agent of leprosy. Macrophages (Mφs) are key players involved in the pathogenesis of leprosy. In this study, immunohistochemical analysis was performed to examine the phenotype of Mφ subpopulations, namely M1, M2, and M4, in the skin lesions of patients diagnosed with leprosy. Based on the database of treatment-naïve patients treated between 2015 and 2019 at the Department of Dermatology of the University of the State of Pará, Belém, routine clinical screening samples were identified. The monolabeling protocol was used for M1 macrophages (iNOS, IL-6, TNF-α) and M2 macrophages (IL-10, IL-13, CD163, Arginase 1, TGF-β, FGFb), and the double-labeling protocol was used for M4 macrophages (IL-6, MMP7, MRP8, TNF-α e CD68). To confirm the M4 macrophage lineage, double labeling of the monoclonal antibodies CD68 and MRP8 was also performed. Our results demonstrated a statistically significant difference for the M1 phenotype among the Virchowian (VV) (4.5 ± 1.3, p < 0.0001), Borderline (1.6 ± 0.4, p < 0.0001), and tuberculoid (TT) (12.5 ± 1.8, p < 0.0001) clinical forms of leprosy. Additionally, the M2 phenotype showed a statistically significant difference among the VV (12.5 ± 2.3, p < 0.0001), Borderline (1.3 ± 0.2, p < 0.0001), and TT (3.2 ± 0.7, p < 0.0001) forms. For the M4 phenotype, a statistically significant difference was observed in the VV (9.8 ± 1.7, p < 0.0001), Borderline (1.2 ± 0.2, p < 0.0001), and TT (2.6 ± 0.7, p < 0.0001) forms. A significant correlation was observed between the VV M1 and M4 (r = 0.8712; p = 0.0000) and between the VV M2 × TT M1 (r = 0.834; p = 0.0002) phenotypes. The M1 Mφs constituted the predominant Mφ subpopulation in the TT and Borderline forms of leprosy, whereas the M2 Mφs showed increased immunoexpression and M4 was the predominant Mφ phenotype in VV leprosy. These results confirm the relationship of the Mφ profile with chronic pathological processes of the inflammatory response in leprosy.
Collapse
Affiliation(s)
- Tatiane Costa Quaresma
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
| | - Lívia de Aguiar Valentim
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
| | - Jorge Rodrigues de Sousa
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
| | - Tinara Leila de Souza Aarão
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
- School of Medicine, Federal University of Para-UFPA, Altamira 68440-000, Brazil
| | - Hellen Thais Fuzii
- Health Department, Tropical Medicine Center, Federal University of Para-NMT-UFPA, Belem 66055-240, Brazil
| | | | - Juarez de Souza
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
| | - Juarez Antônio Simões Quaresma
- Health Department, Center for Biological and Health Sciences, State University of Para-CCBS, UEPA, Belem 66087-662, Brazil
- School of Medicine, Federal University of Para-UFPA, Altamira 68440-000, Brazil
- Health Department, Tropical Medicine Center, Federal University of Para-NMT-UFPA, Belem 66055-240, Brazil
- School of Medicine, Sao Paulo University, Sao Paulo 01246-903, Brazil
| |
Collapse
|
17
|
Duan H, Wang L, Huangfu M, Li H. The impact of microbiota-derived short-chain fatty acids on macrophage activities in disease: Mechanisms and therapeutic potentials. Biomed Pharmacother 2023; 165:115276. [PMID: 37542852 DOI: 10.1016/j.biopha.2023.115276] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/07/2023] Open
Abstract
Short-chain fatty acids (SCFAs) derived from the fermentation of carbohydrates by gut microbiota play a crucial role in regulating host physiology. Among them, acetate, propionate, and butyrate are key players in various biological processes. Recent research has revealed their significant functions in immune and inflammatory responses. For instance, butyrate reduces the development of interferon-gamma (IFN-γ) generating cells while promoting the development of regulatory T (Treg) cells. Propionate inhibits the initiation of a Th2 immune response by dendritic cells (DCs). Notably, SCFAs have an inhibitory impact on the polarization of M2 macrophages, emphasizing their immunomodulatory properties and potential for therapeutics. In animal models of asthma, both butyrate and propionate suppress the M2 polarization pathway, thus reducing allergic airway inflammation. Moreover, dysbiosis of gut microbiota leading to altered SCFA production has been implicated in prostate cancer progression. SCFAs trigger autophagy in cancer cells and promote M2 polarization in macrophages, accelerating tumor advancement. Manipulating microbiota- producing SCFAs holds promise for cancer treatment. Additionally, SCFAs enhance the expression of hypoxia-inducible factor 1 (HIF-1) by blocking histone deacetylase, resulting in increased production of antibacterial effectors and improved macrophage-mediated elimination of microorganisms. This highlights the antimicrobial potential of SCFAs and their role in host defense mechanisms. This comprehensive review provides an in-depth analysis of the latest research on the functional aspects and underlying mechanisms of SCFAs in relation to macrophage activities in a wide range of diseases, including infectious diseases and cancers. By elucidating the intricate interplay between SCFAs and macrophage functions, this review aims to contribute to the understanding of their therapeutic potential and pave the way for future interventions targeting SCFAs in disease management.
Collapse
Affiliation(s)
- Hongliang Duan
- Department of Thyroid Surgery, the Second Hospital of Jilin University, Changchun 130000, China
| | - LiJuan Wang
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun 130000, China.
| | - Mingmei Huangfu
- Department of Thyroid Surgery, the Second Hospital of Jilin University, Changchun 130000, China
| | - Hanyang Li
- Department of Endocrinology, the Second Hospital of Jilin University, Changchun 130000, China
| |
Collapse
|
18
|
Jiang B, Luo Y, Yan N, Shen Z, Li W, Hou C, Xiao L, Ma C, Zhang L, Chen Y, Cheng X, Lian M, Ji C, Zhu Z, Wang Z. An X-ray inactivated vaccine against Pseudomonas aeruginosa Keratitis in mice. Vaccine 2023:S0264-410X(23)00627-8. [PMID: 37353454 DOI: 10.1016/j.vaccine.2023.05.066] [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: 11/14/2022] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/25/2023]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is one of the most prevalent pathogens of bacterial keratitis. Bacterial keratitis is a major cause of blindness worldwide. The rising incidence of multidrug resistance of P. aeruginosa precludes treatment with conventional antibiotics. Herein, we evaluated the protective efficiency and explored the possible underlying mechanism of an X-ray inactivated vaccine (XPa) using a murine P. aeruginosa keratitis model. Mice immunized with XPa exhibit reduced corneal bacterial loads and pathology scores. XPa vaccination induced corneal macrophage polarization toward M2, averting an excessive inflammatory reaction. Furthermore, histological observations indicated that XPa vaccination suppressed corneal fibroblast activation and prevented irreversible visual impairment. The potency of XPa against keratitis highlights its potential utility as an effective and promising vaccine candidate for P. aeruginosa.
Collapse
Affiliation(s)
- Boguang Jiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Yingjie Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Naihong Yan
- Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhixue Shen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Wenfang Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Chen Hou
- Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lirong Xiao
- Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Cuicui Ma
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Li Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Yanwei Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Xingjun Cheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Mao Lian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Chengjie Ji
- Department of Laboratory Medicine, The People's Hospital of Jianyang City, Chengdu 641400, China
| | - Ziyi Zhu
- Department of Laboratory Medicine, The People's Hospital of Jianyang City, Chengdu 641400, China
| | - Zhenling Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China.
| |
Collapse
|
19
|
Zhou Q, Liu J, Yan J, Guo Z, Zhang F. Magnetic microspheres mimicking certain functions of macrophages: Towards precise antibacterial potency for bone defect healing. Mater Today Bio 2023; 20:100651. [PMID: 37206878 PMCID: PMC10189291 DOI: 10.1016/j.mtbio.2023.100651] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/27/2023] [Accepted: 04/29/2023] [Indexed: 05/21/2023] Open
Abstract
A variety of novel biomaterials have recently been developed to promote bone regeneration. However, the current biomaterials cannot accurately and effectively resist bacterial invasion. In this study, we constructed microspheres that mimic certain functions of macrophages as additives to bone repair materials, which can be manipulated as demanded to resist bacteria effectively and protect bone defect healing. Firstly, we prepared gelatin microspheres (GMSs) by an emulsion-crosslinking method, which were subsequently coated with polydopamine (PDA). Then, amino antibacterial nanoparticles obtained by a nanoprecipitation-self-assembly method and commercial amino magnetic nanoparticles were modified onto these PDA-coated GMSs to construct the functionalized microspheres (FMSs). The results showed that the FMSs possessed a rough topography and could be manipulated by a 100-400 mT static magnetic field to migrate directionally in unsolidified hydrogels. Moreover, in vitro experiments with near-infrared (NIR) showed that the FMSs had a sensitive and recyclable photothermal performance and could capture and kill Porphyromonas gingivalis by releasing reactive oxygen species. Finally, the FMSs were mixed with osteogenic hydrogel precursor, injected into the Sprague-Dawley rat periodontal bone defect of maxillary first molar (M1), and subsequently driven by magnetism to the cervical surface of M1 and the outer surface of the gel system for targeted sterilization under NIR, thus protecting the bone defect healing. In conclusion, the FMSs had excellent manipulation and antimicrobial performances. This provided us with a promising strategy to construct light-magnetism-responsive antibacterial materials to build a beneficial environment for bone defect healing.
Collapse
Affiliation(s)
- Qiao Zhou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
| | - Jun Liu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, 210008, China
| | - Jia Yan
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
| | - Zhaobin Guo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Feimin Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
- Corresponding author. Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University; Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China.
| |
Collapse
|
20
|
Zhao T, Zhang Z, Li Y, Sun Z, Liu L, Deng X, Guo J, Zhu D, Cao S, Chai Y, Nikolaevna UV, Maratbek S, Wang Z, Zhang H. Brucella abortus modulates macrophage polarization and inflammatory response by targeting glutaminases through the NF-κB signaling pathway. Front Immunol 2023; 14:1180837. [PMID: 37325614 PMCID: PMC10266586 DOI: 10.3389/fimmu.2023.1180837] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/05/2023] [Indexed: 06/17/2023] Open
Abstract
Objectives The mechanism of Brucella infection regulating macrophage phenotype has not been completely elucidated until now. This study aimed to determine the mechanism of Brucella abortus in the modulation of macrophage phenotype using RAW264.7 cells as a model. Materials and methods RT-qPCR, ELISA and flow cytometry were used to detect the inflammatory factor production and phenotype conversion associated with M1/M2 polarization of macrophages by Brucella abortus infection. Western blot and immunofluorescence were used to analyze the role of nuclear factor kappa B (NF-κB) signaling pathway in regulation of Brucella abortus-induced macrophage polarization. Chromatin immunoprecipitation sequencing (Chip-seq), bioinformatics analysis and luciferase reporter assay were used to screen and validate NF-κB target genes associated with macrophage polarization and further verify its function. Results The results demonstrate that B. abortus induces a macrophage phenotypic switch and inflammatory response in a time-dependent manner. With the increase of infection time, B. abortus infection-induced M1-type increased first, peaked at 12 h, and then decreased, whereas the M2-type decreased first, trough at 12 h, and then increased. The trend of intracellular survival of B. abortus was consistent with that of M2 type. When NF-κB was inhibited, M1-type polarization was inhibited and M2-type was promoted, and the intracellular survival of B. abortus increased significantly. Chip-seq and luciferase reporter assay results showed that NF-κB binds to the glutaminase gene (Gls). Gls expression was down-regulated when NF-κB was inhibited. Furthermore, when Gls was inhibited, M1-type polarization was inhibited and M2-type was promoted, the intracellular survival of B. abortus increased significantly. Our data further suggest that NF-κB and its key target gene Gls play an important role in controlling macrophage phenotypic transformation. Conclusions Taken together, our study demonstrates that B. abortus infection can induce dynamic transformation of M1/M2 phenotype in macrophages. Highlighting NF-κB as a central pathway that regulates M1/M2 phenotypic transition. This is the first to elucidate the molecular mechanism of B. abortus regulation of macrophage phenotype switch and inflammatory response by regulating the key gene Gls, which is regulated by the transcription factor NF-κB.
Collapse
Affiliation(s)
- Tianyi Zhao
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Zedan Zhang
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Yitao Li
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Zhihua Sun
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Liangbo Liu
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Xingmei Deng
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Jia Guo
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Dexin Zhu
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Shuzhu Cao
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Yingjin Chai
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Usevich Vera Nikolaevna
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
- College of Veterinary, Ural State Agricultural University, Yekaterinburg, Russia
| | - Suleimenov Maratbek
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
- College of Veterinary, National Agricultural University of Kazakhstan, Nur Sultan, Kazakhstan
| | - Zhen Wang
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Hui Zhang
- State International Joint Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, Shihezi, China
| |
Collapse
|
21
|
Xie B, Zhao H, Zhang R, Ding Y, Gao C, He Y, Wang R. Bacteria-mimetic nanomedicine for targeted eradication of intracellular MRSA. J Control Release 2023; 357:371-378. [PMID: 37030543 DOI: 10.1016/j.jconrel.2023.03.053] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/26/2023] [Accepted: 03/30/2023] [Indexed: 04/10/2023]
Abstract
Drug-resistant infections caused by intracellular bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA), which are often hidden inside macrophages, pose a significant threat to human health. Various nanomedicines have been developed to combat intracellular MRSA; however, their poor uptake and fast clearance from macrophages often result in insufficient enrichment of antibacterial agents intracellularly, leading to low antibacterial efficacy. Here, we developed bacterial membrane-coated mesoporous SiO2 nanoparticles (MSN) loaded with vancomycin (Van), a classic antibiotic. These nanoparticles can be specifically recognized and internalized by macrophages and self-aggregated into micron-sized MSN clusters based on cucurbit[7]uril-adamantane host-guest interactions, allowing for slow clearance and extended retention in infected macrophages. The acid-triggered, sustainable release of Van from MSN aggregates effectively killed MRSA in infected macrophages and significantly alleviated inflammation caused by intracellular bacterial infections both in vitro and in vivo. This work not only provides a practical solution to effectively treat drug-resistant intracellular infections but also offers new insights for the design and development of antibacterial nanomaterials.
Collapse
Affiliation(s)
- Beibei Xie
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, SAR 999078, PR China; Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, 401331 Shapingba, Chongqing, PR China
| | - Huichao Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, SAR 999078, PR China
| | - Ruixue Zhang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, 401331 Shapingba, Chongqing, PR China
| | - Yuanfu Ding
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, SAR 999078, PR China
| | - Cheng Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, SAR 999078, PR China
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, 55 Daxuecheng South Road, 401331 Shapingba, Chongqing, PR China.
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, SAR 999078, PR China.
| |
Collapse
|
22
|
Azevedo MDCS, Marques H, Binelli LS, Malange MSV, Devides AC, Fachin LRV, Soares CT, Belone ADFF, Rosa PS, Garlet GP, Trombone APF. B lymphocytes deficiency results in altered immune response and increased susceptibility to Mycobacterium leprae in a murine leprosy model. Cytokine 2023; 165:156184. [PMID: 36996537 DOI: 10.1016/j.cyto.2023.156184] [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: 11/27/2022] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023]
Abstract
Leprosy is a chronic and infectious disease that primarily affects the skin and peripheral nervous system, presenting a wide spectrum of clinical forms with different degrees of severity. The distinct host immune response patters developed in the response to the bacillus Mycobacterium leprae, the leprosy etiologic agent, are associated with the spectral clinical forms and outcome of the disease. In this context, B cells are allegedly involved in the disease immunopathogenesis, usually as antibody-producing cells, but also as potential effector or regulatory elements. In order to determine the regulatory B cells role in experimental leprosy, this study evaluated the outcome of M. leprae infection in B cell deficient mice (BKO) and WT C57Bl/6 control, by means of microbiological/bacilloscopic, immunohistochemical and molecular analysis, performed 8 months after M. leprae inoculation. The results demonstrated that infected BKO showed a higher bacilli number when compared with WT animals, demonstrating the importance of these cells in experimental leprosy. The molecular analysis demonstrates that the expression of IL-4, IL-10 and TGF-β was significantly higher in the BKO footpads when compared to WT group. Conversely, there was no difference in IFN-γ, TNF-α and IL-17 expression levels in BKO and WT groups. IL-17 expression was significantly higher in the lymph nodes of WT group. The immunohistochemical analysis revealed that M1 (CD80+) cells counts were significantly lower in the BKO group, while no significant difference was observed to M2 (CD206+) counts, resulting a skewed M1/M2 balance. These results demonstrated that the absence of B lymphocytes contribute to the persistence and multiplication of M. leprae, probably due to the increased expression of the IL-4, IL-10 and TGF-β cytokines, as well as a decrease in the number of M1 macrophages in the inflammatory site.
Collapse
Affiliation(s)
| | - Heloísa Marques
- Centro Universitário Sagrado Coração, UNISAGRADO - Bauru, SP, Brazil; Universidade Federal do Piauí - Parnaíba, PI, Brazil
| | - Larissa S Binelli
- Centro Universitário Sagrado Coração, UNISAGRADO - Bauru, SP, Brazil
| | | | - Amanda C Devides
- Centro Universitário Sagrado Coração, UNISAGRADO - Bauru, SP, Brazil
| | | | | | | | | | - Gustavo P Garlet
- Bauru School of Dentistry, Sao Paulo University - FOB/USP - Bauru, SP, Brazil
| | | |
Collapse
|
23
|
Zhao D, Feng W, Kang X, Li H, Liu F, Zheng W, Li G, Wang X. Dual-targeted poly(amino acid) nanoparticles deliver drug combinations on-site: an intracellular synergistic strategy to eliminate intracellular bacteria. J Mater Chem B 2023; 11:2958-2971. [PMID: 36919349 DOI: 10.1039/d3tb00125c] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Multi-drug combinations are a common strategy for the treatment of intracellular bacterial infections. However, different internalized pathways and the accumulation of the composite drugs at different subcellular organelles very much reduce their efficacy. Herein, an intracellular synergistic strategy is proposed, which is realized by on-site delivery of a drug combination using a macrophage/intracellular bacterium-dual targeted drug delivery system (DDS). The DDS is fabricated by encapsulating vancomycin (Van) and curcumin (Cur) into poly(α-N-acryloyl-phenylalanine)-block-poly(β-N-acryloyl-D-aminoalanine-co-2-O-acetyl-α-D-mannosyloxy) nanoparticles, denoted by (Van + Cur)@F(AM) NPs. Mannose ligands on (Van + Cur)@F(AM) NPs trigger their specific internalization in macrophages, while aminoalanine moieties subsequently drive the NPs to target intracellular methicillin-resistant Staphylococcus aureus (MRSA). Thereafter, Van and Cur are durably released in a synergistic dose at the residence site of intracellular MRSA. Under this intracellular synergistic effect, (Van + Cur)@F(AM) NPs show superior elimination efficiency in vitro and in vivo compared to the control groups, including free Van, (Van + Cur), the DDS encapsulated Van and the DDSs separately-encapsulated Van and Cur. Furthermore, (Van + Cur)@F(AM) NPs significantly enhance the in vivo antibacterial capacity by modulating the immune response. Therefore, this dual-targeted DDS-assisted intracellular synergistic antibacterial strategy of drug combination is an effective therapeutic against intracellular bacteria.
Collapse
Affiliation(s)
- Dongdong Zhao
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Wenli Feng
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Xiaoxu Kang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Haofei Li
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Fang Liu
- Department of Oncology of Integrative Chinese and Western Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Weitao Zheng
- Hubei Provincial Key Laboratory of Industrial Microbiology, Sino-German Biomedical Center, National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan, 430068, Hubei Province, China
| | - Guofeng Li
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| | - Xing Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China.
| |
Collapse
|
24
|
Maimaiti Z, Li Z, Xu C, Fu J, Hao LB, Chen JY, Chai W. Host Immune Regulation in Implant-Associated Infection (IAI): What Does the Current Evidence Provide Us to Prevent or Treat IAI? Bioengineering (Basel) 2023; 10:356. [PMID: 36978747 PMCID: PMC10044746 DOI: 10.3390/bioengineering10030356] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/25/2023] [Accepted: 03/03/2023] [Indexed: 03/17/2023] Open
Abstract
The number of orthopedic implants for bone fixation and joint arthroplasty has been steadily increasing over the past few years. However, implant-associated infection (IAI), a major complication in orthopedic surgery, impacts the quality of life and causes a substantial economic burden on patients and societies. While research and study on IAI have received increasing attention in recent years, the failure rate of IAI has still not decreased significantly. This is related to microbial biofilms and their inherent antibiotic resistance, as well as the various mechanisms by which bacteria evade host immunity, resulting in difficulties in diagnosing and treating IAIs. Hence, a better understanding of the complex interactions between biofilms, implants, and host immunity is necessary to develop new strategies for preventing and controlling these infections. This review first discusses the challenges in diagnosing and treating IAI, followed by an extensive review of the direct effects of orthopedic implants, host immune function, pathogenic bacteria, and biofilms. Finally, several promising preventive or therapeutic alternatives are presented, with the hope of mitigating or eliminating the threat of antibiotic resistance and refractory biofilms in IAI.
Collapse
Affiliation(s)
- Zulipikaer Maimaiti
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhuo Li
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Chi Xu
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Jun Fu
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Li-Bo Hao
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Ji-Ying Chen
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| | - Wei Chai
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, Beijing 100048, China
- Department of Orthopaedics, The First Medical Centre, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
25
|
Single-cell transcriptomics of immune cells in lymph nodes reveals their composition and alterations in functional dynamics during the early stages of bubonic plague. SCIENCE CHINA. LIFE SCIENCES 2023; 66:110-126. [PMID: 35943690 DOI: 10.1007/s11427-021-2119-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 04/26/2022] [Indexed: 02/04/2023]
Abstract
Bubonic plague caused by Yersinia pestis is highly infectious and often fatal. Characterization of the host immune response and its subsequent suppression by Y. pestis is critical to understanding the pathogenesis of Y. pestis. Here, we utilized single-cell RNA sequencing to systematically profile the transcriptomes of immune cells in draining lymph nodes (dLNs) during the early stage of Y. pestis infection. Dendritic cells responded to Y. pestis within 2 h post-infection (hpi), followed by the activation of macrophages/monocytes (Mφs/Mons) and recruitment of polymorphonuclear neutrophils (PMNs) to dLNs at 24 hpi. Analysis of cell-to-cell communication suggests that PMNs may be recruited to lymph nodes following the secretion of CCL9 by Mφs/Mons stimulated through CCR1-CCL9 interaction. Significant functional suppression of all the three innate immune cell types occurred during the early stage of infection. In summary, we present a dynamic immune landscape, at single-cell resolution, of murine dLNs involved in the response to Y. pestis infection, which may facilitate the understanding of the plague pathogenesis of during the early stage of infection.
Collapse
|
26
|
Qu M, Zhu H, Zhang X. Extracellular vesicle-mediated regulation of macrophage polarization in bacterial infections. Front Microbiol 2022; 13:1039040. [PMID: 36619996 PMCID: PMC9815515 DOI: 10.3389/fmicb.2022.1039040] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
Extracellular vesicles (EVs) are nanoscale membrane-enveloped vesicles secreted by prokaryotic and eukaryotic cells, which are commonly defined as membrane vesicles (MVs) and exosomes, respectively. They play critical roles in the bacteria-bacteria and bacteria-host interactions. In infectious diseases caused by bacteria, as the first line of defense against pathogens, the macrophage polarization mode commonly determines the success or failure of the host's response to pathogen aggression. M1-type macrophages secrete pro-inflammatory factors that support microbicidal activity, while alternative M2-type macrophages secrete anti-inflammatory factors that perform an antimicrobial immune response but partially allow pathogens to replicate and survive intracellularly. Membrane vesicles (MVs) released from bacteria as a distinctive secretion system can carry various components, including bacterial effectors, nucleic acids, or lipids to modulate macrophage polarization in host-pathogen interaction. Similar to MVs, bacteria-infected macrophages can secrete exosomes containing a variety of components to manipulate the phenotypic polarization of "bystander" macrophages nearby or long distance to differentiate into type M1 or M2 to regulate the course of inflammation. Exosomes can also repair tissue damage associated with the infection by upregulating the levels of anti-inflammatory factors, downregulating the pro-inflammatory factors, and regulating cellular biological behaviors. The study of the mechanisms by which EVs modulate macrophage polarization has opened new frontiers in delineating the molecular machinery involved in bacterial pathogenesis and challenges in providing new strategies for diagnosis and therapy.
Collapse
Affiliation(s)
- Mingjuan Qu
- School of Life Sciences, Ludong University, Yantai, China,Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai, China
| | - Hongwei Zhu
- School of Life Sciences, Ludong University, Yantai, China,Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai, China,Shandong Provincial Key Laboratory of Quality Safety Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Xingxiao Zhang
- School of Life Sciences, Ludong University, Yantai, China,Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai, China,Shandong Breeding Environmental Control Engineering Laboratory, Yantai, China,*Correspondence: Xingxiao Zhang, ✉
| |
Collapse
|
27
|
Zhao QY, Li QH, Fu YY, Ren CE, Jiang AF, Meng YH. Decidual macrophages in recurrent spontaneous abortion. Front Immunol 2022; 13:994888. [PMID: 36569856 PMCID: PMC9781943 DOI: 10.3389/fimmu.2022.994888] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Recurrent spontaneous abortion (RSA) is defined as two or more pregnancy loss, affecting the happiness index of fertility couples. The mechanisms involved in the occurrence of RSA are not clear to date. The primary problem for the maternal immune system is how to establish and maintain the immune tolerance to the semi-allogeneic fetuses. During the pregnancy, decidual macrophages mainly play an important role in the immunologic dialogue. The purpose of this study is to explore decidual macrophages, and to understand whether there is a connection between these cells and RSA by analyzing their phenotypes and functions. Pubmed, Web of Science and Embase were searched. The eligibility criterion for this review was evaluating the literature about the pregnancy and macrophages. Any disagreement between the authors was resolved upon discussion and if required by the judgment of the corresponding author. We summarized the latest views on the phenotype, function and dysfunction of decidual macrophages to illuminate its relationship with RSA.
Collapse
Affiliation(s)
| | | | | | | | | | - Yu-Han Meng
- Center of Reproductive Medicine, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
28
|
Athapaththu AMGK, Lee KT, Kavinda MHD, Lee S, Kang S, Lee MH, Kang CH, Choi YH, Kim GY. Pinostrobin ameliorates lipopolysaccharide (LPS)-induced inflammation and endotoxemia by inhibiting LPS binding to the TLR4/MD2 complex. Biomed Pharmacother 2022; 156:113874. [DOI: 10.1016/j.biopha.2022.113874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/03/2022] [Accepted: 10/13/2022] [Indexed: 11/02/2022] Open
|
29
|
Xiao G, Zhang S, Zhang L, Liu S, Li G, Ou M, Zeng X, Wang Z, Zhang G, Lu S. Untargeted metabolomics analysis reveals Mycobacterium tuberculosis strain H37Rv specifically induces tryptophan metabolism in human macrophages. BMC Microbiol 2022; 22:249. [PMID: 36253713 PMCID: PMC9575276 DOI: 10.1186/s12866-022-02659-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/27/2022] [Indexed: 11/10/2022] Open
Abstract
Background Tuberculosis (TB) caused by Mycobacterium tuberculosis (M. tb) remains a global health issue. The characterized virulent M. tb H37Rv, avirulent M. tb H37Ra and BCG strains are widely used as reference strains to investigate the mechanism of TB pathogenicity. Here, we attempted to determine metabolomic signatures associated with the Mycobacterial virulence in human macrophages through comparison of metabolite profile in THP-1-derived macrophages following exposure to the M. tb H37Rv, M. tb H37Ra and BCG strains. Results Our findings revealed remarkably changed metabolites in infected macrophages compared to uninfected macrophages. H37Rv infection specifically induced 247 differentially changed metabolites compared to H37Ra or BCG infection. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed H37Rv specifically induces tryptophan metabolism. Moreover, quantitative PCR (qPCR) results showed that indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase 2 (TDO2) which converts the tryptophan to a series of biologically second metabolites were up-regulated in H37Rv-infected macrophages compared to H37Ra- or BCG-infected macrophages, confirming the result of enhanced tryptophan metabolism induced by H37Rv infection. These findings indicated that targeting tryptophan (Trp) metabolism may be a potential therapeutic strategy for pulmonary TB. Conclusions We identified a number of differentially changed metabolites that specifically induced in H37Rv infected macrophages. These signatures may be associated with the Mycobacterial virulence in human macrophages. The present findings provide a better understanding of the host response associated with the virulence of the Mtb strain. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-022-02659-y.
Collapse
Affiliation(s)
- Guohui Xiao
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518112, China
| | - Su Zhang
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518112, China
| | - Like Zhang
- School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China
| | - Shuyan Liu
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518112, China
| | - Guobao Li
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518112, China
| | - Min Ou
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518112, China
| | - Xuan Zeng
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518112, China
| | - Zhaoqin Wang
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518112, China.
| | - Guoliang Zhang
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518112, China. .,School of Basic Medical Sciences, Guangdong Medical University, Dongguan, China.
| | - Shuihua Lu
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518112, China.
| |
Collapse
|
30
|
He S, Wang C, Huang Y, Lu S, Li W, Ding N, Chen C, Wu Y. Chlamydia psittaci plasmid-encoded CPSIT_P7 induces macrophage polarization to enhance the antibacterial response through TLR4-mediated MAPK and NF-κB pathways. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119324. [PMID: 35809864 DOI: 10.1016/j.bbamcr.2022.119324] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/23/2022] [Accepted: 06/30/2022] [Indexed: 06/15/2023]
Abstract
Although the protective effects of Chlamydia psittaci plasmid-encoded protein CPSIT_P7 as vaccine antigens to against chlamydial infection have been confirmed in our previous study, the function and mechanism of CPSIT_P7 inducing innate immunity in the antibacterial response remain unknown. Here, we found that plasmid protein CPSIT_P7 could induce M1 macrophage polarization upregulating the genes of the surface molecule CD86, proinflammatory cytokines (TNF-α, IL-6, and IL-1β), and antibacterial effector NO synthase 2 (iNOS). During M1 macrophage polarization, macrophages acquire phagocytic and microbicidal competence, which promotes the host antibacterial response. As we observed that CPSIT_P7-induced M1 macrophages could partially reduce the infected mice pulmonary Chlamydia psittaci load. Furthermore, CPSIT_P7 induced M1 macrophage polarization through the TLR4-mediated MAPK and NF-κB pathways. Collectively, our results highlight the effect of CPSIT_P7 on macrophage polarization and provide new insights into new prevention and treatment strategies for chlamydial infection.
Collapse
Affiliation(s)
- Siqin He
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan 421001, China
| | - Chuan Wang
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan 421001, China
| | - Yanru Huang
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan 421001, China
| | - Simin Lu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan 421001, China
| | - Weiwei Li
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan 421001, China
| | - Nan Ding
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan 421001, China
| | - Chaoqun Chen
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan 421001, China.
| | - Yimou Wu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
31
|
He F, Qiu Y, Wu X, Xia Y, Yang L, Wu C, Li P, Zhang R, Fang R, Li N, Peng Y. Slc6a13 Deficiency Attenuates Pasteurella multocida Infection-Induced Inflammation via Glycine-Inflammasome Signaling. J Innate Immun 2022; 15:107-121. [PMID: 35797984 PMCID: PMC10643921 DOI: 10.1159/000525089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 05/07/2022] [Indexed: 11/19/2022] Open
Abstract
We have previously demonstrated that Slc6a13-deficient (Slc6a13-/-; KO) mice are resistant to P. multocida infection, which might be in connection with macrophage-mediated inflammation; however, the specific metabolic mechanism is still enigmatic. Here we reproduce the less sensitive to P. multocida infection in overall survival assays as well as reduced bacterial loads, tissue lesions, and inflammation of lungs in KO mice. The transcriptome sequencing analysis of wild-type (WT) and KO mice shows a large number of differentially expressed genes that are enriched in amino acid metabolism by functional analysis. Of note, glycine levels are substantially increased in the lungs of KO mice with or without P. multocida infection in comparison to the WT controls. Interestingly, exogenous glycine supplementation alleviates P. multocida infection-induced inflammation. Mechanistically, glycine reduces the production of inflammatory cytokines in macrophages by blocking the activation of inflammasome (NALP1, NLRP3, NLRC4, AIM2, and Caspase-1). Together, Slc6a13 deficiency attenuates P. multocida infection through lessening the excessive inflammatory responses of macrophages involving glycine-inflammasome signaling.
Collapse
Affiliation(s)
- Fang He
- College of Veterinary Medicine, Southwest University, Chongqing, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yangyang Qiu
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xiaoyan Wu
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yaoyao Xia
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Liu Yang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Chenlu Wu
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Pan Li
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Rui Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Rendong Fang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Nengzhang Li
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yuanyi Peng
- College of Veterinary Medicine, Southwest University, Chongqing, China
| |
Collapse
|
32
|
Tseng CC, Huang SY, Tsai HP, Wu CW, Hsieh TH. HDAC6 is a prognostic biomarker that mediates IL-13 expression to regulate macrophage polarization through AP-1 in oral squamous cell carcinoma. Sci Rep 2022; 12:10513. [PMID: 35732647 PMCID: PMC9217956 DOI: 10.1038/s41598-022-14052-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/31/2022] [Indexed: 01/17/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a common malignant tumor worldwide that is characterized by abnormal lesions or malignant hyperplasia of soft and hard tissues in the oral cavity. Previous research has found that HDAC6 may be a potential therapeutic target for cancer patients and has the ability to regulate immune cells. However, the mechanism of HDAC6 in OSCC pathogenesis is unclear. We collected clinical samples and analyzed the level of HDAC6 in OSCC patients. The results showed that in the high HDAC6 expression group, HDAC6 expression was positively correlated with the grade of OSCC (R = 0.182, P = 0.036) and that this group had a 3.248-fold increase in the mortality risk compared with the low HDAC6 expression group (P = 0.003). Survival analysis also identified a correlation between the expression of HDAC6 and overall survival in OSCC patients, and it was found that the expression of HDAC6 was inversely correlated with survival (P ≤ 0.001). In addition, we found that HDAC6 induced IL-13 expression through AP-1, resulting in M2 polarization of macrophages. Together, these results demonstrate that the level of HDAC6 may be a useful prognostic biomarker and offer a novel immune cell-related therapeutic strategy of targeting IL-13 in OSCC.
Collapse
Affiliation(s)
- Chung-Chih Tseng
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan.,Department of Dentistry, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung, 81342, Taiwan
| | - Shi-Ying Huang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, 361021, China
| | - Hung-Pei Tsai
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chia-Wei Wu
- Department of Medical Research, E-Da Hospital/E-Da Cancer Hospital, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Tsung-Hua Hsieh
- Department of Medical Research, E-Da Hospital/E-Da Cancer Hospital, I-Shou University, Kaohsiung, 82445, Taiwan.
| |
Collapse
|
33
|
dos Santos CC, Walburg KV, van Veen S, Wilson LG, Trufen CEM, Nascimento IP, Ottenhoff THM, Leite LCC, Haks MC. Recombinant BCG-LTAK63 Vaccine Candidate for Tuberculosis Induces an Inflammatory Profile in Human Macrophages. Vaccines (Basel) 2022; 10:vaccines10060831. [PMID: 35746439 PMCID: PMC9227035 DOI: 10.3390/vaccines10060831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 11/29/2022] Open
Abstract
Tuberculosis (TB) is one of the top 10 leading causes of death worldwide. The recombinant BCG strain expressing the genetically detoxified A subunit of the thermolabile toxin from Escherichia coli (LTAK63) adjuvant (rBCG-LTAK63) has previously been shown to confer superior protection and immunogenicity compared to BCG in a murine TB infection model. To further investigate the immunological mechanisms induced by rBCG-LTAK63, we evaluated the immune responses induced by rBCG-LTAK63, BCG, and Mycobacterium tuberculosis (Mtb) H37Rv strains in experimental infections of primary human M1 and M2 macrophages at the transcriptomic and cytokine secretion levels. The rBCG-LTAK63-infected M1 macrophages more profoundly upregulated interferon-inducible genes such as IFIT3, OAS3, and antimicrobial gene CXCL9 compared to BCG, and induced higher levels of inflammatory cytokines such as IL-12(p70), TNF-β, and IL-15. The rBCG-LTAK63-infected M2 macrophages more extensively upregulated transcripts of inflammation-related genes, TAP1, GBP1, SLAMF7, TNIP1, and IL6, and induced higher levels of cytokines related to inflammation and tissue repair, MCP-3 and EGF, as compared to BCG. Thus, our data revealed an important signature of immune responses induced in human macrophages by rBCG-LTAK63 associated with increased inflammation, activation, and tissue repair, which may be correlated with a protective immune response against TB.
Collapse
Affiliation(s)
- Carina C. dos Santos
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil;
- Programa de Pós-Graduação Interunidades em Biotecnologia, Universidade de São Paulo, São Paulo 05508-900, Brazil
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (K.V.W.); (S.v.V.); (L.G.W.); (T.H.M.O.); (M.C.H.)
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador 40170-115, Brazil
- Correspondence: (C.C.d.S.); (L.C.C.L.)
| | - Kimberley V. Walburg
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (K.V.W.); (S.v.V.); (L.G.W.); (T.H.M.O.); (M.C.H.)
| | - Suzanne van Veen
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (K.V.W.); (S.v.V.); (L.G.W.); (T.H.M.O.); (M.C.H.)
| | - Louis G. Wilson
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (K.V.W.); (S.v.V.); (L.G.W.); (T.H.M.O.); (M.C.H.)
| | | | - Ivan P. Nascimento
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil;
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (K.V.W.); (S.v.V.); (L.G.W.); (T.H.M.O.); (M.C.H.)
| | - Luciana C. C. Leite
- Laboratório de Desenvolvimento de Vacinas, Instituto Butantan, São Paulo 05503-900, Brazil;
- Programa de Pós-Graduação Interunidades em Biotecnologia, Universidade de São Paulo, São Paulo 05508-900, Brazil
- Correspondence: (C.C.d.S.); (L.C.C.L.)
| | - Mariëlle C. Haks
- Department of Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (K.V.W.); (S.v.V.); (L.G.W.); (T.H.M.O.); (M.C.H.)
| |
Collapse
|
34
|
Gao Y, Jin H, Tan H, Cai X, Sun Y. Erythrocyte-derived extracellular vesicles aggravate inflammation by promoting the proinflammatory macrophage phenotype through TLR4-MyD88-NF-κB-MAPK pathway. J Leukoc Biol 2022; 112:693-706. [PMID: 35411633 DOI: 10.1002/jlb.3a0821-451rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 03/12/2022] [Indexed: 12/31/2022] Open
Abstract
Transfusion of stored erythrocytes is associated with the increased risk of morbidity and mortality in critical infections, but the mechanism is incompletely understood. Previous studies have suggested that RBC-derived extracellular vesicles (EVs) may be potential risk factors for the occurrence of transfusion-related immunomodulation. The purpose of our study was to evaluate the effects of RBC-derived EVs under inflammatory conditions and explore the underlying mechanisms. In vivo, the activity of EVs was evaluated in cecal ligation and puncture (CLP)-induced sepsis. Our results showed that EVs significantly aggravated the inflammatory response to sepsis in serum and lung tissue by promoting the production of the proinflammatory factors tumor necrosis factor-α (TNF-α)-interleukin-6(IL-6), and interleukin-1β (IL-1β) and reduced the survival rate of septic mice in vivo. Importantly, adoptive transfer of EVs-pretreated bone marrow-derived macrophages (BMDMs) obviously aggravated systemic proinflammatory factors in mice after CLP surgery. In vitro, the proinflammatory properties of EVs were shown to elevate TNF-α, IL-6, and IL-1β levels in lipopolysaccharide (LPS)-stimulated BMDMs. Moreover, EVs promoted LPS-induced macrophage polarization into a proinflammatory phenotype. The underlying mechanism might involve EV-mediated up-regulation of TLR4-MyD88-NF-κB-MAPK activity to favor macrophage cytokine production.
Collapse
Affiliation(s)
- Yuhan Gao
- Department of Blood Transfusion, Peking University People's Hospital, Beijing, China
| | - Haiqiang Jin
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Hui Tan
- Department of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen, China
| | - Xiaodong Cai
- Department of Neurosurgery, Shenzhen Second People's Hospital, Shenzhen, China
| | - Yongan Sun
- Department of Neurology, Peking University First Hospital, Beijing, China
| |
Collapse
|
35
|
Zhang R, Yu B, Tian Y, Pang L, Xu T, Cong H, Shen Y. Diversified antibacterial modification and latest applications of polysaccharide-based hydrogels for wound healthcare. APPLIED MATERIALS TODAY 2022; 26:101396. [DOI: 10.1016/j.apmt.2022.101396] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
36
|
Gayer FA, Reichardt SD, Bohnenberger H, Engelke M, Reichardt HM. Characterization of testicular macrophagesubpopulations in mice. Immunol Lett 2022; 243:44-52. [PMID: 35149127 DOI: 10.1016/j.imlet.2022.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/25/2022] [Accepted: 02/07/2022] [Indexed: 12/11/2022]
Abstract
Testis is an immune privileged site, a feature that prevents germ cells from eliciting an autoimmune response. Macrophages contribute to this state of tolerance by adopting an immunoregulatory phenotype. Here, we further characterized their features in mice by analyzing surface markers, anatomic localization as well as morphology and function. Testicular macrophages (TMF) were stained for various surface receptors, and MHCII and CD206 were found to be most suitable to discriminate between two subpopulations. Our immunohistochemical analysis further confirmed a predominant localization of CD206+ cells in the interstitial space. Imaging flow cytometry revealed that both subtypes of TMF differed in size and contrast, and to some extent also in their ability to engulf high-molecular dextran. To investigate whether the polarization of the immune system had any influence on the phenotype of TMF, we compared C57BL/6 and BALB/c mice. Importantly, our analysis revealed that the abundance of cells expressing either MHCII or any of the scavenger receptors CD206, CD163 and CD71 differed between both mouse strains. In addition, the presence of the glucocorticoid receptor in macrophages affected the ratio between individual subpopulations, which is consistent with a crucial role of glucocorticoids in macrophage polarization. Collectively, our results indicate that TMF are composed in a variable ratio of distinct subsets with characteristic features, which may shape the immune privilege of the testis also in humans.
Collapse
Affiliation(s)
- Fabian A Gayer
- University Medical Center Göttingen, Institute for Cellular and Molecular Immunology, Göttingen, Germany; University Medical Center Göttingen, Clinic of Urology, Göttingen, Germany
| | - Sybille D Reichardt
- University Medical Center Göttingen, Institute for Cellular and Molecular Immunology, Göttingen, Germany
| | | | - Michael Engelke
- University Medical Center Göttingen, Institute for Cellular and Molecular Immunology, Göttingen, Germany
| | - Holger M Reichardt
- University Medical Center Göttingen, Institute for Cellular and Molecular Immunology, Göttingen, Germany.
| |
Collapse
|
37
|
Kulkarni RR, Gaghan C, Mohammed J. Avian Macrophage Responses to Virulent and Avirulent Clostridium perfringens. Pathogens 2022; 11:pathogens11010100. [PMID: 35056048 PMCID: PMC8778324 DOI: 10.3390/pathogens11010100] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 11/16/2022] Open
Abstract
The present study evaluated the avian macrophage responses against Clostridium perfringens that varied in their ability to cause necrotic enteritis in chickens. Strains CP5 (avirulent-netB+), CP1 (virulent-netB+), and CP26 (highly virulent-netB+tpeL+) were used to evaluate their effect on macrophages (MQ-NCSU cells) and primary splenic and cecal tonsil mononuclear cells. The bacilli (whole cells) or their secretory products from all three strains induced a significant increase in the macrophage transcription of Toll-like receptor (TLR)21, TLR2, interleukin (IL)-1β, inducible nitric oxide synthase (iNOS), and CD80 genes as well as their nitric oxide (NO) production and major histocompatibility complex (MHC)-II surface expression compared to an unstimulated control. The CP1 and CP26-induced expression of interferon (IFN)γ, IL-6, CD40 genes, MHC-II upregulation, and NO production was significantly higher than that of CP5 and control groups. Furthermore, splenocytes and cecal tonsillocytes stimulated with bacilli or secretory products from all the strains showed a significant increase in the frequency of macrophages, their surface expression of MHC-II and NO production, while CP26-induced responses were significantly higher for the rest of the groups. In summary, macrophage interaction with C. perfringens can lead to cellular activation and, the ability of this pathogen to induce macrophage responses may depend on its level of virulence.
Collapse
|
38
|
Linking the antimicrobial and anti-inflammatory effects of immortelle essential oil with its chemical composition - The interplay between the major and minor constituents. Food Chem Toxicol 2021; 158:112666. [PMID: 34762977 DOI: 10.1016/j.fct.2021.112666] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/23/2021] [Accepted: 11/04/2021] [Indexed: 11/21/2022]
Abstract
Immortelle (Helichrysum italicum, Asteraceae) essential oil has been widely used in alternative medicine to accelerate wound healing, as well as in cosmetic products to stimulate skin regeneration and to reduce the appearance of wrinkles. It is also considered a natural and safe culinary spice that could also be applied in the food industry as a preservative in the future. The therapeutic efficacy of this oil changes with the natural variability of the composition. Herein we tested and mutually compared the antimicrobial and anti-inflammatory activities of four commercial immortelle oils differing in the relative amounts of marker compounds, i.e. neryl esters, α-pinene, γ- and ar-curcumenes, and β-diketones. The anti-inflammatory effect of selected chromatographic fractions, enriched in the aforementioned constituents, was evaluated by studying toxicity toward rat peritoneal macrophages, their nitric oxide production, myeloperoxidase, and arginase activities. Subsequently, the compositional and activity data were subjected to a multivariate statistical treatment to reveal the possible correlation(s) between the percentage of essential-oil constituents and the observed activities. The obtained results imply that immortelle oil efficiency as an antimicrobial and/or anti-inflammatory agent is most plausibly a result of a synergistic action between its constituents, and/or, rather unexpectedly, the presence of some minor constituents.
Collapse
|
39
|
Gajanayaka N, Dong SXM, Ali H, Iqbal S, Mookerjee A, Lawton DA, Caballero RE, Cassol E, Cameron DW, Angel JB, Crawley AM, Kumar A. TLR-4 Agonist Induces IFN-γ Production Selectively in Proinflammatory Human M1 Macrophages through the PI3K-mTOR- and JNK-MAPK-Activated p70S6K Pathway. THE JOURNAL OF IMMUNOLOGY 2021; 207:2310-2324. [PMID: 34551966 DOI: 10.4049/jimmunol.2001191] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 08/18/2021] [Indexed: 12/18/2022]
Abstract
IFN-γ, a proinflammatory cytokine produced primarily by T cells and NK cells, activates macrophages and engages mechanisms to control pathogens. Although there is evidence of IFN-γ production by murine macrophages, IFN-γ production by normal human macrophages and their subsets remains unknown. Herein, we show that human M1 macrophages generated by IFN-γ and IL-12- and IL-18-stimulated monocyte-derived macrophages (M0) produce significant levels of IFN-γ. Further stimulation of IL-12/IL-18-primed macrophages or M1 macrophages with agonists for TLR-2, TLR-3, or TLR-4 significantly enhanced IFN-γ production in contrast to the similarly stimulated M0, M2a, M2b, and M2c macrophages. Similarly, M1 macrophages generated from COVID-19-infected patients' macrophages produced IFN-γ that was enhanced following LPS stimulation. The inhibition of M1 differentiation by Jak inhibitors reversed LPS-induced IFN-γ production, suggesting that differentiation with IFN-γ plays a key role in IFN-γ induction. We subsequently investigated the signaling pathway(s) responsible for TLR-4-induced IFN-γ production in M1 macrophages. Our results show that TLR-4-induced IFN-γ production is regulated by the ribosomal protein S6 kinase (p70S6K) through the activation of PI3K, the mammalian target of rapamycin complex 1/2 (mTORC1/2), and the JNK MAPK pathways. These results suggest that M1-derived IFN-γ may play a key role in inflammation that may be augmented following bacterial/viral infections. Moreover, blocking the mTORC1/2, PI3K, and JNK MAPKs in macrophages may be of potential translational significance in preventing macrophage-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Niranjala Gajanayaka
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Simon Xin Min Dong
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Hamza Ali
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Faculty of Applied Medical Sciences, Taibah University, Medina, Kingdom of Saudi Arabia
| | - Salma Iqbal
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ananda Mookerjee
- Apoptosis Research Center, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - David A Lawton
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ramon Edwin Caballero
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Apoptosis Research Center, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| | - Edana Cassol
- Department of Biology, Carleton University, Ottawa, Ontario, Canada
| | - Donald William Cameron
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Jonathan B Angel
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Angela M Crawley
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Department of Biology, Carleton University, Ottawa, Ontario, Canada.,Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Center for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ontario, Canada; and
| | - Ashok Kumar
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada; .,Apoptosis Research Center, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada.,Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
40
|
Ali H, Dong SXM, Gajanayaka N, Cassol E, Angel JB, Kumar A. Selective Induction of Cell Death in Human M1 Macrophages by Smac Mimetics Is Mediated by cIAP-2 and RIPK-1/3 through the Activation of mTORC. THE JOURNAL OF IMMUNOLOGY 2021; 207:2359-2373. [PMID: 34561230 DOI: 10.4049/jimmunol.2100108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 08/28/2021] [Indexed: 12/22/2022]
Abstract
Inflammatory macrophages have been implicated in many diseases, including rheumatoid arthritis and inflammatory bowel disease. Therefore, targeting macrophage function and activation may represent a potential strategy to treat macrophage-associated diseases. We have previously shown that IFN-γ-induced differentiation of human M0 macrophages toward proinflammatory M1 state rendered them highly susceptible to the cytocidal effects of second mitochondria-derived activator of caspases mimetics (SMs), antagonist of the inhibitors of apoptosis proteins (IAPs), whereas M0 and anti-inflammatory M2c macrophages were resistant. In this study, we investigated the mechanism governing SM-induced cell death during differentiation into M1 macrophages and in polarized M1 macrophages. IFN-γ stimulation conferred on M0 macrophages the sensitivity to SM-induced cell death through the Jak/STAT, IFN regulatory factor-1, and mammalian target of rapamycin complex-1 (mTORC-1)/ribosomal protein S6 kinase pathways. Interestingly, mTORC-1 regulated SM-induced cell death independent of M1 differentiation. In contrast, SM-induced cell death in polarized M1 macrophages is regulated by the mTORC-2 pathway. Moreover, SM-induced cell death is regulated by cellular IAP (cIAP)-2, receptor-interacting protein kinase (RIPK)-1, and RIPK-3 degradation through mTORC activation during differentiation into M1 macrophages and in polarized M1 macrophages. In contrast to cancer cell lines, SM-induced cell death in M1 macrophages is independent of endogenously produced TNF-α, as well as the NF-κB pathway. Collectively, selective induction of cell death in human M1 macrophages by SMs may be mediated by cIAP-2, RIPK-1, and RIPK-3 degradation through mTORC activation. Moreover, blocking cIAP-1/2, mTORC, or IFN regulatory factor-1 may represent a promising therapeutic strategy to control M1-associated diseases.
Collapse
Affiliation(s)
- Hamza Ali
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ontario, Canada; .,Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada.,Faculty of Applied Medical Sciences, Taibah University, Medina, Kingdom of Saudi Arabia
| | - Simon Xin Min Dong
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Niranjala Gajanayaka
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Edana Cassol
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Jonathan B Angel
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ontario, Canada.,Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Medicine, Faculty of Medicine, University of Ottawa, Ontario, Canada; and
| | - Ashok Kumar
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ontario, Canada; .,Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada.,Department of Pathology and Laboratory Medicine, University of Ottawa, Ontario, Canada
| |
Collapse
|
41
|
Sakama S, Kurusu K, Morita M, Oizumi T, Masugata S, Oka S, Yokomizo S, Nishimura M, Morioka T, Kakinuma S, Shimada Y, Nakamura AJ. An Enriched Environment Alters DNA Repair and Inflammatory Responses After Radiation Exposure. Front Immunol 2021; 12:760322. [PMID: 34745135 PMCID: PMC8570081 DOI: 10.3389/fimmu.2021.760322] [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: 08/18/2021] [Accepted: 10/08/2021] [Indexed: 11/22/2022] Open
Abstract
After the Fukushima Daiichi Nuclear Power Plant accident, there is growing concern about radiation-induced carcinogenesis. In addition, living in a long-term shelter or temporary housing due to disasters might cause unpleasant stress, which adversely affects physical and mental health. It's been experimentally demonstrated that "eustress", which is rich and comfortable, has beneficial effects for health using mouse models. In a previous study, mice raised in the enriched environment (EE) has shown effects such as suppression of tumor growth and enhancement of drug sensitivity during cancer treatment. However, it's not yet been evaluated whether EE affects radiation-induced carcinogenesis. Therefore, to evaluate whether EE suppresses a radiation-induced carcinogenesis after radiation exposure, in this study, we assessed the serum leptin levels, radiation-induced DNA damage response and inflammatory response using the mouse model. In brief, serum and tissues were collected and analyzed over time in irradiated mice after manipulating the raising environment during the juvenile or adult stage. To assess the radiation-induced DNA damage response, we performed immunostaining for phosphorylated H2AX which is a marker of DNA double-strand break. Focusing on the polarization of macrophages in the inflammatory reaction that has an important role in carcinogenesis, we performed analysis using tissue immunofluorescence staining and RT-qPCR. Our data confirmed that EE breeding before radiation exposure improved the responsiveness to radiation-induced DNA damage and basal immunity, further suppressing the chronic inflammatory response, and that might lead to a reduction of the risk of radiation-induced carcinogenesis.
Collapse
Affiliation(s)
- Sae Sakama
- Department of Biological Science, College of Sciences, Ibaraki University, Mito, Japan
| | - Keisuke Kurusu
- Department of Biological Science, College of Sciences, Ibaraki University, Mito, Japan
| | - Mayu Morita
- Department of Biological Science, College of Sciences, Ibaraki University, Mito, Japan
| | - Takashi Oizumi
- Department of Biological Science, College of Sciences, Ibaraki University, Mito, Japan
| | - Shinya Masugata
- Department of Biological Science, College of Sciences, Ibaraki University, Mito, Japan
| | - Shohei Oka
- Department of Biological Science, College of Sciences, Ibaraki University, Mito, Japan
| | - Shinya Yokomizo
- Department of Radiation Effects Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Mayumi Nishimura
- Department of Radiation Effects Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Takamitsu Morioka
- Department of Radiation Effects Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Shizuko Kakinuma
- Department of Radiation Effects Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Yoshiya Shimada
- Executive Director, National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Asako J. Nakamura
- Department of Biological Science, College of Sciences, Ibaraki University, Mito, Japan
| |
Collapse
|
42
|
Long Y, Li L, Xu T, Wu X, Gao Y, Huang J, He C, Ma T, Ma L, Cheng C, Zhao C. Hedgehog artificial macrophage with atomic-catalytic centers to combat Drug-resistant bacteria. Nat Commun 2021; 12:6143. [PMID: 34686676 PMCID: PMC8536674 DOI: 10.1038/s41467-021-26456-9] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 09/30/2021] [Indexed: 02/05/2023] Open
Abstract
Pathogenic drug-resistant bacteria represent a threat to human health, for instance, the methicillin-resistant Staphylococcus aureus (MRSA). There is an ever-growing need to develop non-antibiotic strategies to fight bacteria without triggering drug resistance. Here, we design a hedgehog artificial macrophage with atomic-catalytic centers to combat MRSA by mimicking the “capture and killing” process of macrophages. The experimental studies and theoretical calculations reveal that the synthesized materials can efficiently capture and kill MRSA by the hedgehog topography and substantial generation of •O2− and HClO with its Fe2N6O catalytic centers. The synthesized artificial macrophage exhibits a low minimal inhibition concentration (8 μg/mL Fe-Art M with H2O2 (100 μM)) to combat MRSA and rapidly promote the healing of bacteria-infected wounds on rabbit skin. We suggest that the application of this hedgehog artificial macrophage with “capture and killing” capability and high ROS-catalytic activity will open up a promising pathway to develop antibacterial materials for bionic and non-antibiotic disinfection strategies. The increase in drug-resistant bacteria is a world-wide health issue that demands the development of alternatives to standard antibiotic treatments. In this study, the authors synthesise a hedgehog artificial macrophage with heme-mimetic catalytic centres, and peroxidase- and haloperoxidase-mimicking activities, for the treatment of methicillin-resistant Staphylococcus aureus.
Collapse
Affiliation(s)
- Yanping Long
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, 610065, Chengdu, China
| | - Ling Li
- Department of Ultrasound, West China Hospital, Sichuan University, 610041, Chengdu, China.,Department of Ultrasound, Affiliated Hospital of North Sichuan Medical College, 637000, Nanchong, China
| | - Tao Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, 610065, Chengdu, China
| | - Xizheng Wu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, 610065, Chengdu, China
| | - Yun Gao
- College of Biomass Science and Engineering, Textile Institute, Sichuan University, 610065, Chengdu, China
| | - Jianbo Huang
- Department of Ultrasound, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Chao He
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, 610065, Chengdu, China
| | - Tian Ma
- Department of Ultrasound, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Lang Ma
- Department of Ultrasound, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Chong Cheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, 610065, Chengdu, China.
| | - Changsheng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, 610065, Chengdu, China. .,College of Biomedical Engineering, National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, China. .,College of Chemical Engineering, Sichuan University, 610065, Chengdu, China.
| |
Collapse
|
43
|
Yu Y, Sun B, Wang Z, Yang M, Cui Z, Lin S, Jin M, Yi C. Exosomes From M2 Macrophage Promote Peritendinous Fibrosis Posterior Tendon Injury via the MiR-15b-5p/FGF-1/7/9 Pathway by Delivery of circRNA-Ep400. Front Cell Dev Biol 2021; 9:595911. [PMID: 34513819 PMCID: PMC8432299 DOI: 10.3389/fcell.2021.595911] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 05/24/2021] [Indexed: 01/02/2023] Open
Abstract
Achilles tendon rupture prognosis is usually unsatisfactory. After the tendon is injured, it may not function properly because of the fibrotic healing response, which restrains tendon motion. Inflammatory monocytes and tissue-resident macrophages are indispensable regulators in tissue repair, fibrosis, and regeneration. Exosomes from macrophages are crucial factors in tissue microenvironment regulation following tissue injury. This study therefore aimed to clarify the roles of macrophage exosomes in tendon injury (TI) repair. The results show that macrophages play a role after TI. M1 macrophages were increased relative to peritendinous fibrosis after TI. High-throughput sequencing showed abnormal expression of circular RNAs (circRNAs) between exosomes from M2 and M0 macrophages. Among the abnormal expressions of circRNA, circRNA-Ep400 was significantly increased in M2 macrophage exosomes. The results also show that M2 macrophage-derived circRNA-Ep400-containing exosomes are important for promoting peritendinous fibrosis after TI. Bioinformatics and dual-luciferase reporting experiments confirmed that miR-15b-5p and fibroblast growth factor (FGF)-1/7/9 were downstream targets of circRNA-Ep400. High circRNA-Ep400-containing exosome treatment inhibited miR-15b-5p, but promoted FGF1/7/9 expression in both fibroblasts and tenocytes. Furthermore, high circRNA-Ep400-containing exosome treatment promoted fibrosis, proliferation, and migration in both fibroblasts and tenocytes. Taken together, the results show that M2 macrophage-derived circRNA-Ep400-containing exosomes promote peritendinous fibrosis after TI via the miR-15b-5p/FGF-1/7/9 pathway, which suggests novel therapeutics for tendon injury treatment.
Collapse
Affiliation(s)
- Yinxian Yu
- Department of Orthopaedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Binbin Sun
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuoying Wang
- Department of Orthopaedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengkai Yang
- Department of Orthopaedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi Cui
- Department of Orthopaedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Subin Lin
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Mingming Jin
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Chengqing Yi
- Department of Orthopaedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
44
|
Allen PE, Noland RC, Martinez JJ. Rickettsia conorii survival in THP-1 macrophages involves host lipid droplet alterations and active rickettsial protein production. Cell Microbiol 2021; 23:e13390. [PMID: 34464019 DOI: 10.1111/cmi.13390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 12/29/2022]
Abstract
Rickettsia conorii is a Gram-negative, cytosolic intracellular bacterium that has classically been investigated in terms of endothelial cell infection. However, R. conorii and other human pathogenic Rickettsia species have evolved mechanisms to grow in various cell types, including macrophages, during mammalian infection. During infection of these phagocytes, R. conorii shifts the host cell's overall metabolism towards an anti-inflammatory M2 response, metabolically defined by an increase in host lipid metabolism and oxidative phosphorylation. Lipid metabolism has more recently been identified as a key regulator of host homeostasis through modulation of immune signalling and metabolism. Intracellular pathogens have adapted mechanisms of hijacking host metabolic pathways including host lipid catabolic pathways for various functions required for growth and survival. In the present study, we hypothesised that alterations of host lipid droplets initiated by lipid catabolic pathways during R. conorii infection is important for bacterial survival in macrophages. Herein, we determined that host lipid droplet modulation is initiated early during R. conorii infection, and these alterations rely on active bacteria and lipid catabolic pathways. We also find that these lipid catabolic pathways are essential for efficient bacterial survival. Unlike the mechanisms used by other intracellular pathogens, the catabolism of lipid droplets induced by R. conorii infection is independent of upstream host peroxisome proliferator-activated receptor-alpha (PPARα) signalling. Inhibition of PPARɣ signalling and lipid droplet accumulation in host cells cause a significant decrease in R. conorii survival suggesting a negative correlation with lipid droplet production and R. conorii survival. Together, these results strongly suggest that the modulation of lipid droplets in macrophage cells infected by R. conorii is an important and underappreciated aspect of the infection process. TAKE AWAYS: Host lipid droplets are differentially altered in early and replicative stages of THP-1 macrophage infection with R. conorii. Lipid droplet alterations are initiated in a bacterial-dependent manner and do not require host peroxisome proliferator-activated receptors α or ɣ activation. Pharmacological inhibition of host lipid catabolic processes during R. conorii infection indicates a requirement of lipid catabolism for bacterial survival and initiation of lipid droplet modulation. A significant increase in host lipid droplets during infection has a negative impact on R. conorii survival in THP-1 macrophages.
Collapse
Affiliation(s)
- Paige E Allen
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Robert C Noland
- Skeletal Muscle Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Juan J Martinez
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| |
Collapse
|
45
|
The Evolving Roles of Cardiac Macrophages in Homeostasis, Regeneration, and Repair. Int J Mol Sci 2021; 22:ijms22157923. [PMID: 34360689 PMCID: PMC8347787 DOI: 10.3390/ijms22157923] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Macrophages were first described as phagocytic immune cells responsible for maintaining tissue homeostasis by the removal of pathogens that disturb normal function. Historically, macrophages have been viewed as terminally differentiated monocyte-derived cells that originated through hematopoiesis and infiltrated multiple tissues in the presence of inflammation or during turnover in normal homeostasis. However, improved cell detection and fate-mapping strategies have elucidated the various lineages of tissue-resident macrophages, which can derive from embryonic origins independent of hematopoiesis and monocyte infiltration. The role of resident macrophages in organs such as the skin, liver, and the lungs have been well characterized, revealing functions well beyond a pure phagocytic and immunological role. In the heart, recent research has begun to decipher the functional roles of various tissue-resident macrophage populations through fate mapping and genetic depletion studies. Several of these studies have elucidated the novel and unexpected roles of cardiac-resident macrophages in homeostasis, including maintaining mitochondrial function, facilitating cardiac conduction, coronary development, and lymphangiogenesis, among others. Additionally, following cardiac injury, cardiac-resident macrophages adopt diverse functions such as the clearance of necrotic and apoptotic cells and debris, a reduction in the inflammatory monocyte infiltration, promotion of angiogenesis, amelioration of inflammation, and hypertrophy in the remaining myocardium, overall limiting damage extension. The present review discusses the origin, development, characterization, and function of cardiac macrophages in homeostasis, cardiac regeneration, and after cardiac injury or stress.
Collapse
|
46
|
Wen J, Chen C, Luo M, Liu X, Guo J, Wei T, Gu X, Gu S, Ning Y, Li Y. Notch Signaling Ligand Jagged1 Enhances Macrophage-Mediated Response to Helicobacter pylori. Front Microbiol 2021; 12:692832. [PMID: 34305857 PMCID: PMC8297740 DOI: 10.3389/fmicb.2021.692832] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022] Open
Abstract
Helicobacter pylori (H. pylori) is one of the gram-negative bacteria that mainly colonize the stomach mucosa and cause many gastrointestinal diseases, such as gastritis, peptic ulcer, and gastric cancer. Macrophages play a key role in eradicating H. pylori. Recent data have shown that Notch signaling could modulate the activation and bactericidal activities of macrophages. However, the role of Notch signaling in macrophages against H. pylori remains unclear. In the present study, in the co-culture model of macrophages with H. pylori, the inhibition of Notch signaling using γ-secretase decreased the expression of inducible nitric oxide synthase (iNOS) and its product, nitric oxide (NO), and downregulated the secretion of pro-inflammatory cytokine and attenuated phagocytosis and bactericidal activities of macrophages to H. pylori. Furthermore, we identified that Jagged1, one of Notch signaling ligands, was both upregulated in mRNA and protein level in activated macrophages induced by H. pylori. Clinical specimens showed that the number of Jagged1+ macrophages in the stomach mucosa from H. pylori-infected patients was significantly higher than that in healthy control. The overexpression of Jagged1 promoted bactericidal activities of macrophages against H. pylori and siRNA-Jagged1 presented the opposite effect. Besides, the addition of exogenous rJagged1 facilitated the pro-inflammatory mediators of macrophages against H. pylori, but the treatment of anti-Jagged1 neutralizing antibody attenuated it. Taken together, these results suggest that Jagged1 is a promoting molecule for macrophages against H. pylori, which will provide insight for exploring Jagged1 as a novel therapeutic target for the control of H. pylori infection.
Collapse
Affiliation(s)
- Junjie Wen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Chuxi Chen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Meiqun Luo
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xiaocong Liu
- The First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Jiading Guo
- The First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Tingting Wei
- The First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Xinyi Gu
- The First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Sinan Gu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yunshan Ning
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yan Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| |
Collapse
|
47
|
Franco R, Lillo A, Rivas-Santisteban R, Reyes-Resina I, Navarro G. Microglial Adenosine Receptors: From Preconditioning to Modulating the M1/M2 Balance in Activated Cells. Cells 2021; 10:1124. [PMID: 34066933 PMCID: PMC8148598 DOI: 10.3390/cells10051124] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/19/2021] [Accepted: 05/01/2021] [Indexed: 12/14/2022] Open
Abstract
Neuronal survival depends on the glia, that is, on the astroglial and microglial support. Neurons die and microglia are activated not only in neurodegenerative diseases but also in physiological aging. Activated microglia, once considered harmful, express two main phenotypes: the pro-inflammatory or M1, and the neuroprotective or M2. When neuroinflammation, i.e., microglial activation occurs, it is important to achieve a good M1/M2 balance, i.e., at some point M1 microglia must be skewed into M2 cells to impede chronic inflammation and to afford neuronal survival. G protein-coupled receptors in general and adenosine receptors in particular are potential targets for increasing the number of M2 cells. This article describes the mechanisms underlying microglial activation and analyzes whether these cells exposed to a first damaging event may be ready to be preconditioned to better react to exposure to more damaging events. Adenosine receptors are relevant due to their participation in preconditioning. They can also be overexpressed in activated microglial cells. The potential of adenosine receptors and complexes formed by adenosine receptors and cannabinoids as therapeutic targets to provide microglia-mediated neuroprotection is here discussed.
Collapse
Affiliation(s)
- Rafael Franco
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain;
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Alejandro Lillo
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain;
| | - Rafael Rivas-Santisteban
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain;
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Irene Reyes-Resina
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain;
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Gemma Navarro
- CiberNed, Network Research Center, Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28034 Madrid, Spain;
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain;
| |
Collapse
|
48
|
Dhingra D, Marathe SA, Sharma N, Marathe A, Chakravortty D. Modeling the immune response to Salmonella during typhoid. Int Immunol 2021; 33:281-298. [PMID: 33406267 DOI: 10.1093/intimm/dxab003] [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: 08/12/2020] [Accepted: 01/05/2021] [Indexed: 11/13/2022] Open
Abstract
Several facets of the host immune response to Salmonella infection have been studied independently at great depths to understand the progress and pathogenesis of Salmonella infection. The circumstances under which a Salmonella-infected individual succumbs to an active disease, evolves as a persister or clears the infection are not understood in detail. We have adopted a system-level approach to develop a continuous-time mechanistic model. We considered key interactions of the immune system state variables with Salmonella in the mesenteric lymph node to determine the final disease outcome deterministically and exclusively temporally. The model accurately predicts the disease outcomes and immune response trajectories operational during typhoid. The results of the simulation confirm the role of anti-inflammatory (M2) macrophages as a site for persistence and relapsing infection. Global sensitivity analysis highlights the importance of both bacterial and host attributes in influencing the disease outcome. It also illustrates the importance of robust phagocytic and anti-microbial potential of M1 macrophages and dendritic cells (DCs) in controlling the disease. Finally, we propose therapeutic strategies for both antibiotic-sensitive and antibiotic-resistant strains (such as IFN-γ therapy, DC transfer and phagocytic potential stimulation). We also suggest prevention strategies such as improving the humoral response and macrophage carrying capacity, which could complement current vaccination schemes for enhanced efficiency.
Collapse
Affiliation(s)
- Divy Dhingra
- Department of Mechanical Engineering, Birla Institute of Technology & Science, Pilani, Rajasthan, India
| | - Sandhya Amol Marathe
- Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Rajasthan, India
| | - Nandita Sharma
- Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, Rajasthan, India
| | - Amol Marathe
- Department of Mechanical Engineering, Birla Institute of Technology & Science, Pilani, Rajasthan, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| |
Collapse
|
49
|
Pathological and Immunohistochemical Characteristics of Granuloma and Lymphatics in Cheilitis Granulomatosa. Am J Dermatopathol 2021; 44:83-91. [PMID: 33878045 DOI: 10.1097/dad.0000000000001952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Cheilitis granulomatosa (CG) is an idiopathic, rare, and chronic granulomatous disorder involving the lips. We characterized the pathological and immunohistopathological findings of these granulomas and their relationship with the lymphatic vessels. Pathologically confirmed cases of primary CG from 2001 to 2016 were collected. Cases of inflammatory cheilitis without the presence of granuloma were included in the control group. Demographic data, clinical presentation, response to therapy, and pathological differences were compared. Periodic acid-Schiff and acid-fast stains excluded patients having infections. CD68, CD163, and D2-40 stains demonstrated features of granuloma, macrophage polarization, and the relationship between granuloma and lymphatic vessels. Thirteen patients diagnosed with CG were enrolled. Thirteen people were enrolled in the control group. The granulomas were either mononuclear or sarcoidal. They were predominantly positive for CD68 but negative for CD163. Perilymphatic granulomas were found in all patients. Intralymphatic histiocytosis and lymphatic dilatation were more commonly observed in patients diagnosed with CG than those in controls (54% vs. 15%, P = 0.03 and 92% vs. 23%, P < 0.01). TH1 immune response due to CD68+ M1 macrophages results in CG. Perilymphatic aggregation of macrophages and intralymphatic histiocytosis were important pathological clues for diagnosis.
Collapse
|
50
|
Kavelaars A, Heijnen CJ. T Cells as Guardians of Pain Resolution. Trends Mol Med 2021; 27:302-313. [PMID: 33431239 PMCID: PMC8005447 DOI: 10.1016/j.molmed.2020.12.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/20/2020] [Accepted: 12/10/2020] [Indexed: 01/08/2023]
Abstract
Despite successful research efforts aimed at understanding pain mechanisms, there is still no adequate treatment for many patients suffering from chronic pain. The contribution of neuroinflammation to chronic pain is widely acknowledged. Here, we summarize findings indicating that T cells play a key role in the suppression of pain. An active contribution of the immune system to resolution of pain may explain why immunosuppressive drugs are often not sufficient to control pain. This would also imply that dysregulation of certain immune functions promote transition to chronic pain. Conversely, stimulating the endogenous immune-mediated resolution pathways may provide a potent approach to treat chronic pain.
Collapse
Affiliation(s)
- Annemieke Kavelaars
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas, M.D. Anderson Cancer Center, Zayed Building, M.D. Anderson Boulevard, Houston, TX 77030, USA.
| | - Cobi J Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas, M.D. Anderson Cancer Center, Zayed Building, M.D. Anderson Boulevard, Houston, TX 77030, USA.
| |
Collapse
|