1
|
Wu J, Yu G, Zhang X, Staiger MP, Gupta TB, Yao H, Wu X. A fructan-type garlic polysaccharide upregulates immune responses in macrophage cells and in immunosuppressive mice. Carbohydr Polym 2024; 344:122530. [PMID: 39218552 DOI: 10.1016/j.carbpol.2024.122530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/20/2024] [Accepted: 07/20/2024] [Indexed: 09/04/2024]
Abstract
The anti-inflammatory effects of plant polysaccharides are well known. However, the stimulatory effects of polysaccharides under immunosuppressive conditions and their link with the polysaccharide structure is underexplored. In this work, the immune modulatory effects of a garlic polysaccharide (GP) are investigated via in vitro and vivo methods. It is observed that GP enhance the immune response of macrophages (RAW264.7) as indicated by the elevated levels of nitric oxide, TNF-α and IL-6. The observation that GP are able to stimulate the immune response in vitro was then explored with the use of an immunosuppressed mouse model. Surprisingly, GP exhibited dose-dependent up-regulatory impacts on the cyclophosphamide (CTX) suppressed levels of cytokines such as IFN-γ and IL-6 and immunoglobulins (e.g. IgA and IgG). The GP intervention reversed histopathological damage to the small intestine and spleen and increased fecal short-chain fatty acid levels. Moreover, GP modulates the gut microbiota dysbiosis by increasing the abundance of immunogenic bacteria such as g__norank_f__Erysipelotrichaceae, while inhibiting the over-abundance of g_Bacteroides. Functional predictions indicated that gut biomarkers of GP possessed the functions of glycoside hydrolase family 32 (GH32) and β-fructofuranosidase. It is concluded that GP is a promising immunostimulant for immune-compromised individuals.
Collapse
Affiliation(s)
- Junfeng Wu
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China; Department of Mechanical Engineering, University of Canterbury, Private Bag 4800, 8140 Christchurch, New Zealand
| | - Guoxing Yu
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Xiaosa Zhang
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Mark P Staiger
- Department of Mechanical Engineering, University of Canterbury, Private Bag 4800, 8140 Christchurch, New Zealand
| | - Tanushree B Gupta
- Food System Integrity Team, Hopkirk Research Institute, AgResearch Ltd, Palmerston North 4474, New Zealand
| | - Hong Yao
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China.
| | - Xiyang Wu
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
2
|
Medhasi S, Sangphech N, Permpalung N, Torvorapanit P, Plongla R, Worasilchai N. Functional characterization of macrophages and change of Th1/Th2 balance in patients with pythiosis after Pythium insidiosum antigen immunotherapy. Sci Rep 2024; 14:27363. [PMID: 39521871 PMCID: PMC11550834 DOI: 10.1038/s41598-024-78756-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
There has been limited research into the role of the Pythium insidiosum antigen (PIA) in modulating immune response in patients with pythiosis. This study investigated the balance of T helper type 2 (Th2) and T helper type 1 (Th1) responses after receiving PIA immunotherapy in patients with pythiosis. Next, the phagocytic activity and phagocytic index of IFN-γ primed PIA-treated macrophages were examined. Furthermore, the phagocytosis of infective P. insidiosum zoospores by macrophages was investigated. This work showed that the PIA vaccine induced Th1 response and M1 macrophages in patients with vascular pythiosis who survived and those with localized pythiosis. Phagocytic activity and phagocytic index were increased considerably in localized pythiosis patients compared to vascular pythiosis patients with hematological diseases. IFN-γ priming of PIA-treated macrophages against P. insidiosum zoospores enhanced the phagocytic activity and phagocytic index in vascular and localized pythiosis patients. Macrophages engulfed P. insidiosum zoospores, but the zoospores continued germination, resulting in macrophage death. Overall, our results suggest that PIA can modulate the immune responses, contributing to higher levels of Th1-type cytokine and potentially improving the survival of patients with vascular pythiosis. This study is the first to uncover that P. insidiosum zoospores can survive within macrophages.
Collapse
Affiliation(s)
- Sadeep Medhasi
- Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, Phayathai Road, Pathumwan, Bangkok, 10330, Thailand
- Research Unit of Medical Mycology Diagnosis, Chulalongkorn University, Bangkok, Thailand
| | - Naunpun Sangphech
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, Thailand
| | - Nitipong Permpalung
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pattama Torvorapanit
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Thai Red Cross Emerging Infectious Diseases Clinical Center, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Rongpong Plongla
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Navaporn Worasilchai
- Department of Transfusion Medicine and Clinical Microbiology, Faculty of Allied Health Sciences, Chulalongkorn University, Phayathai Road, Pathumwan, Bangkok, 10330, Thailand.
- Research Unit of Medical Mycology Diagnosis, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
3
|
Moon S, Jung M, Go S, Hong J, Sohn HS, Kim C, Kang M, Lee BJ, Kim J, Lim J, Kim BS. Engineered Nanoparticles for Enhanced Antitumoral Synergy Between Macrophages and T Cells in the Tumor Microenvironment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410340. [PMID: 39252658 DOI: 10.1002/adma.202410340] [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: 07/17/2024] [Revised: 08/22/2024] [Indexed: 09/11/2024]
Abstract
T cells and macrophages have the potential to collaborate to eliminate tumor cells efficiently. Macrophages can eliminate tumor cells through phagocytosis and subsequently activate T cells by presenting tumor antigens. The activated T cells, in turn, can kill tumor cells and redirect tumor-associated macrophages toward an antitumoral M1 phenotype. However, checkpoint molecules expressed on tumor cells impede the collaborative action of these immune cells. Meanwhile, monotherapy with a single immune checkpoint inhibitor (ICI) for either macrophages or T cells yields suboptimal efficacy in cancer patients. To address this challenge, here a nanoparticle capable of efficiently delivering dual ICIs to tumors for both macrophages and T cells is developed. These programmed cell death protein 1 (PD-1)-transfected macrophage membrane-derived nanoparticles (PMMNPs) can target tumors and provide signal-regulatory protein alpha and PD-1 to block CD47 and programmed cell death-ligand 1 (PD-L1), respectively, on tumor cells. PMMNPs enhance macrophage-mediated cancer cell phagocytosis and antigen presentation, promote T cell activation, and induce the reprogramming of macrophages toward an antitumoral phenotype. In syngeneic tumor-bearing mice, PMMNPs demonstrate superior therapeutic efficacy compared to nanoparticles delivering single ICIs and non-targeted delivery of anti-CD47 and anti-PD-L1 antibodies. PMMNPs capable of augmenting the antitumoral interplay between macrophages and T cells may offer a promising avenue for cancer immunotherapy.
Collapse
Affiliation(s)
- Sangjun Moon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Mungyo Jung
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seokhyeong Go
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hee Su Sohn
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cheesue Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Mikyung Kang
- School of Health and Environmental Science, Korea University, Seoul, 02841, Republic of Korea
| | - Byung Joon Lee
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jungwoo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jinwoong Lim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Engineering Research, Institute of Chemical Processes, and BioMAX, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
4
|
da Silveira BP, Cohen ND, Lawhon SD, Watson RO, Bordin AI. Protective immune response against Rhodococcus equi: An innate immunity-focused review. Equine Vet J 2024. [PMID: 39258739 DOI: 10.1111/evj.14214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/30/2024] [Indexed: 09/12/2024]
Abstract
Rhodococcus equi causes pyogranulomatous pneumonia in foals and immunocompromised people. Despite decades of research efforts, no vaccine is available against this common cause of disease and death in foals. The purpose of this narrative review is to summarise the current understanding of interactions between R. equi and the host innate immune system, to describe features of the immune response that are associated with resistance or susceptibility to R. equi infection, and help guide strategies for developing novel approaches for preventing R. equi infections. Virulence of R. equi in foals has been attributed to the virulence associated protein A which allows intracellular survival in macrophages by preventing acidification of R. equi-containing vacuole. Additionally, foal susceptibility to R. equi infection is associated with immaturity and naivety of innate and adaptive immune systems, while adult horses with fully functional immune system are resistant to pneumonia. Specific interaction between R. equi and innate immune cells can result in bacterial survival or death; learning how to manipulate these responses to control infection is critical to prevent pneumonia in foals. Administration of live vaccines and stimulation of innate immune responses appears to improve foals' immune response and has the potential to overcome the challenges of foal active vaccination and elicit protection against pneumonia.
Collapse
Affiliation(s)
- Bibiana Petri da Silveira
- Equine Infectious Disease Laboratory, Department of Large Animal Clinical Sciences, Texas A&M University, School of Veterinary Medicine & Biomedical Sciences, College Station, Texas, USA
| | - Noah D Cohen
- Equine Infectious Disease Laboratory, Department of Large Animal Clinical Sciences, Texas A&M University, School of Veterinary Medicine & Biomedical Sciences, College Station, Texas, USA
| | - Sara D Lawhon
- Department of Veterinary Pathobiology, Texas A&M University, School of Veterinary Medicine & Biomedical Sciences, College Station, Texas, USA
| | - Robert O Watson
- Department of Microbial Pathogenesis & Immunology, Texas A&M University, School of Medicine, College Station, Texas, USA
| | - Angela I Bordin
- Equine Infectious Disease Laboratory, Department of Large Animal Clinical Sciences, Texas A&M University, School of Veterinary Medicine & Biomedical Sciences, College Station, Texas, USA
| |
Collapse
|
5
|
Liu Z, Zhang X, Xiong S, Huang S, Ding X, Xu M, Yao J, Liu S, Zhao F. Endothelial dysfunction of syphilis: Pathogenesis. J Eur Acad Dermatol Venereol 2024; 38:1478-1490. [PMID: 38376088 DOI: 10.1111/jdv.19899] [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: 08/28/2023] [Accepted: 01/19/2024] [Indexed: 02/21/2024]
Abstract
Treponema pallidum is the causative factor of syphilis, a sexually transmitted disease (STD) characterized by perivascular infiltration of inflammatory cells, vascular leakage, swelling and proliferation of endothelial cells (ECs). The endothelium lining blood and lymphatic vessels is a key barrier separating body fluids from host tissues and is a major target of T. pallidum. In this review, we focus on how T. pallidum establish intimate interactions with ECs, triggering endothelial dysfunction such as endothelial inflammation, abnormal repairment and damage of ECs. In addition, we summarize that migration and invasion of T. pallidum across vascular ECs may occur through two pathways. These two mechanisms of transendothelial migration are paracellular and cholesterol-dependent, respectively. Herein, clarifying the relationship between T. pallidum and endothelial dysfunction is of great significance to provide novel strategies for diagnosis and prevention of syphilis, and has a great potential prospect of clinical application.
Collapse
Affiliation(s)
- Zhaoping Liu
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Xiaohong Zhang
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Shun Xiong
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Shaobin Huang
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Xuan Ding
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Man Xu
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Jiangchen Yao
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Shuangquan Liu
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| | - Feijun Zhao
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
6
|
Gay L, Madariaga Zarza S, Abou Atmeh P, Rouvière MS, Andrieu J, Richaud M, Boumaza A, Miquel L, Diallo AB, Bechah Y, Otmani Idrissi M, La Scola B, Olive D, Resseguier N, Bretelle F, Mezouar S, Mege JL. Protective role of macrophages from maternal-fetal interface in unvaccinated coronavirus disease 2019 pregnant women. J Med Virol 2024; 96:e29819. [PMID: 39030992 DOI: 10.1002/jmv.29819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/29/2024] [Accepted: 07/02/2024] [Indexed: 07/22/2024]
Abstract
Pregnant women represent a high-risk population for Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) infection. The presence of SARS-CoV-2 has been reported in placenta from infected pregnant women, but whether the virus influences placenta immune response remains unclear. We investigated the properties of maternal-fetal interface macrophages (MFMs) in a cohort of unvaccinated women who contracted coronavirus disease 2019 (COVID-19) during their pregnancy. We reported an infiltration of CD163+ macrophages in placenta from COVID-19 women 19 whereas lymphoid compartment was not affected. Isolated MFMs exhibited nonpolarized activated signature (NOS2, IDO1, IFNG, TNF, TGFB) mainly in women infected during the second trimester of pregnancy. COVID-19 during pregnancy primed MFM to produce type I and III interferon response to SARS-CoV-2 (Wuhan and δ strains), that were unable to elicit this in MFMs from healthy pregnant women. COVID-19 also primed SARS-CoV-2 internalization by MFM in an angiotensin-converting enzyme 2-dependent manner. Activation and recall responses of MFMs were influenced by fetal sex. Collectively, these findings support a role for MFMs in the local immune response to SARS-CoV-2 infection, provide a basis for protective placental immunity in COVID-19, and highlight the interest of vaccination in pregnant women.
Collapse
Affiliation(s)
- Laetitia Gay
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
| | - Sandra Madariaga Zarza
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
| | - Perla Abou Atmeh
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
| | - Marie-Sarah Rouvière
- Institut Paoli-Calmettes, UM105, Centre National de la Recherche Scientifique, Aix-Marseille University, Marseille, France
| | - Jonatane Andrieu
- Centre National de la Recherche Scientifique, Etablissement Français du Sang, Anthropologie bio-culturelle, Droit, Ethique et Santé, Aix-Marseille University, Marseille, France
| | - Manon Richaud
- Institut Paoli-Calmettes, UM105, Centre National de la Recherche Scientifique, Aix-Marseille University, Marseille, France
| | - Asma Boumaza
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
| | - Laura Miquel
- Department of Gynaecology-Obstetrics, La Conception Hospital, Marseille, France
| | - Aïssatou Bailo Diallo
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
| | - Yassina Bechah
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
| | - Myriem Otmani Idrissi
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
| | - Bernard La Scola
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
| | - Daniel Olive
- Institut Paoli-Calmettes, UM105, Centre National de la Recherche Scientifique, Aix-Marseille University, Marseille, France
| | - Noémie Resseguier
- Assistance Publique-Hôpitaux de Marseille, La Timone Hospital, Department of Epidemiology and Health Economics, Clinical Research Unit, Direction of Health Research, Aix Marseille University, Marseille, France
| | - Florence Bretelle
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
- Department of Gynaecology-Obstetrics, La Conception Hospital, Marseille, France
| | - Soraya Mezouar
- Centre National de la Recherche Scientifique, Etablissement Français du Sang, Anthropologie bio-culturelle, Droit, Ethique et Santé, Aix-Marseille University, Marseille, France
| | - Jean-Louis Mege
- Institut Recherche Développement, Assistance Publique-Hôpitaux de Marseille, Microbe, Evolution, Phylogeny and Infection, Aix-Marseille University, Marseille, France
- Department of Immunology, Timone Hospital, Marseille, France
| |
Collapse
|
7
|
Chang J, Gao X, Yang F, Qiang P, Fan L, Liu Z, Shimosawa T, Xu Q, Chang Y. Esaxerenone Inhibits Interferon-γ Induced Pyroptosis of Macrophages in the Lungs of Aldosterone-treated Mice. Inflammation 2024:10.1007/s10753-024-02030-5. [PMID: 38713304 DOI: 10.1007/s10753-024-02030-5] [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: 03/19/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 05/08/2024]
Abstract
Lung immune cells such as lymphocytes and macrophages can induce an inflammatory response due to the activation of mineralocorticoid receptor (MR), which is manifested by the infiltration of inflammatory cells and the secretion of inflammatory cytokines and subsequent apoptosis, pyroptosis and necrosis of intrinsic lung cells and immune cells. Macrophages are immune cells that are abundant in the lung and act as the first line of defense against pathogens but are also aggravating factors of infection. The activation of the renin-angiotensin-aldosterone system (RAAS), especially aldosterone-stimulated MR activation, can induce macrophage and CD8+ T cell aggregation and the secretion of cytokines such as tumor necrosis factor-α (TNF-α) and interferon-gamma (IFN-γ). Increased IFN-γ secretion can induce macrophage pyroptosis and the release of interleukin 1-β (IL-1β), aggravating lung injury. In this study, lung injury in C57BL/6 mice was induced by subcutaneous micro-osmotic pump infusion of aldosterone. After 12 weeks of administration, the kidney, heart, blood vessels and lungs all showed obvious inflammatory injury, which manifested as rapid accumulation of macrophages. The overexpression of IFN-γ in the lungs of aldosterone-treated mice and the stimulation of MH-S and RAW264.7 alveolar macrophages (AMs) with aldosterone in vitro showed that IFN-γ induced pyroptosis of macrophages via the activation of the inflammasome, and the MR blocker esaxerenone effectively inhibited this effect and alleviated lung injury. In addition, IFN-γ secreted by CD8+ T cells is associated with macrophage pyroptosis. In conclusion, the inhibition of macrophage pyroptosis can effectively alleviate lung injury.
Collapse
Affiliation(s)
- Jingyue Chang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Xiaomeng Gao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Fan Yang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
- Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Panpan Qiang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
- Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Lili Fan
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Ziqian Liu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Tatsuo Shimosawa
- Department of Clinical Laboratory, School of Medicine, International University of Health and Welfare, Narita, 286-8686, Japan
| | - Qingyou Xu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China.
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China.
- Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China.
| | - Yi Chang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China.
- Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China.
| |
Collapse
|
8
|
Bestion E, Rachid M, Tijeras-Raballand A, Roth G, Decaens T, Ansaldi C, Mezouar S, Raymond E, Halfon P. Ezurpimtrostat, A Palmitoyl-Protein Thioesterase-1 Inhibitor, Combined with PD-1 Inhibition Provides CD8 + Lymphocyte Repopulation in Hepatocellular Carcinoma. Target Oncol 2024; 19:95-106. [PMID: 38133710 DOI: 10.1007/s11523-023-01019-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Palmitoyl-protein thioesterase-1 (PPT1) is a clinical stage druggable target for inhibiting autophagy in cancer. OBJECTIVE We aimed to determine the cellular and molecular activity of targeting PPT1 using ezurpimtrostat, in combination with an anti-PD-1 antibody. METHODS In this study we used a transgenic immunocompetent mouse model of hepatocellular carcinoma. RESULTS Herein, we revealed that inhibition of PPT1 using ezurpimtrostat decreased the liver tumor burden in a mouse model of hepatocellular carcinoma by inducing the penetration of lymphocytes into tumors when combined with anti-programmed death-1 (PD-1). Inhibition of PPT1 potentiates the effects of anti-PD-1 immunotherapy by increasing the expression of major histocompatibility complex (MHC)-I at the surface of liver cancer cells and modulates immunity through recolonization and activation of cytotoxic CD8+ lymphocytes. CONCLUSIONS Ezurpimtrostat turns cold tumors into hot tumors and, thus, could improve T cell-mediated immunotherapies in liver cancer.
Collapse
Affiliation(s)
- Eloïne Bestion
- Genoscience Pharma, 10, Rue d'Iéna, 13006, Marseille, France
| | - Madani Rachid
- Genoscience Pharma, 10, Rue d'Iéna, 13006, Marseille, France
| | | | - Gael Roth
- Centre hospitalouniversitaire Grenoble Alpes/Institute for Advanced Biosciences, Centre national de la recherché scienti-fique, Unité mixte de recherche 5309-Institut national de la santé et de la recherche médicale U1209, University Grenoble Alpes/Hepato-Gastroenterology and Digestive Oncology Department, 38043, Grenoble, France
| | - Thomas Decaens
- Centre hospitalouniversitaire Grenoble Alpes/Institute for Advanced Biosciences, Centre national de la recherché scienti-fique, Unité mixte de recherche 5309-Institut national de la santé et de la recherche médicale U1209, University Grenoble Alpes/Hepato-Gastroenterology and Digestive Oncology Department, 38043, Grenoble, France
| | | | - Soraya Mezouar
- Genoscience Pharma, 10, Rue d'Iéna, 13006, Marseille, France
- Etablissement français du sang, Centre national de la recherche scientifique, Anthropologie bio-culturelle, droit, éthique et santé, "Biologie des Groupes Sanguins", Aix-Marseille University, Marseille, France
| | - Eric Raymond
- Genoscience Pharma, 10, Rue d'Iéna, 13006, Marseille, France
- Oncology Department, Groupe Hospitalier Paris Saint Joseph, Paris, France
| | - Philippe Halfon
- Genoscience Pharma, 10, Rue d'Iéna, 13006, Marseille, France.
| |
Collapse
|
9
|
Wu A, Wang Y, Ali A, Xu Z, Zhang D, Zhumanov K, Sheng J, Yi J. Design of a multi-epitope vaccine against brucellosis fused to IgG-fc by an immunoinformatics approach. Front Vet Sci 2023; 10:1238634. [PMID: 37937155 PMCID: PMC10625910 DOI: 10.3389/fvets.2023.1238634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/27/2023] [Indexed: 11/09/2023] Open
Abstract
Introduction Brucella, a type of intracellular Gram-negative bacterium, has unique features and acts as a zoonotic pathogen. It can lead to abortion and infertility in animals. Eliminating brucellosis becomes very challenging once it spreads among both humans and animals, putting a heavy burden on livestock and people worldwide. Given the increasing spread of brucellosis, it is crucial to develop improved vaccines for susceptible animals to reduce the disease's impact. Methods In this study, we effectively used an immunoinformatics approach with advanced computer software to carefully identify and analyze important antigenic parts of Brucella abortus. Subsequently, we skillfully designed chimeric peptides to enhance the vaccine's strength and effectiveness. We used computer programs to find four important parts of the Brucella bacteria that our immune system recognizes. Then, we carefully looked for eight parts that are recognized by a type of white blood cell called cytotoxic T cells, six parts recognized by T helper cells, and four parts recognized by B cells. We connected these parts together using a special link, creating a strong new vaccine. To make the vaccine even better, we added some extra parts called molecular adjuvants. These included something called human β-defensins 3 (hBD-3) that we found in a database, and another part that helps the immune system called PADRE. We attached these extra parts to the beginning of the vaccine. In a new and clever way, we made the vaccine even stronger by attaching a part from a mouse's immune system to the end of it. This created a new kind of vaccine called MEV-Fc. We used advanced computer methods to study how well the MEV-Fc vaccine interacts with certain receptors in the body (TLR-2 and TLR-4). Results In the end, Immunosimulation predictions showed that the MEV-Fc vaccine can make the immune system respond strongly, both in terms of cells and antibodies. Discussion In summary, our results provide novel insights for the development of Brucella vaccines. Although further laboratory experiments are required to assess its protective effect.
Collapse
Affiliation(s)
- Aodi Wu
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Yueli Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Adnan Ali
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Zhenyu Xu
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Dongsheng Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Kairat Zhumanov
- College of Veterinary Medicine, Kazakhstan Kazakh State Agricultural University, Almaty, Kazakhstan
| | - Jinliang Sheng
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| | - Jihai Yi
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
10
|
Sheng G, Chu H, Duan H, Wang W, Tian N, Liu D, Sun H, Sun Z. LRRC25 Inhibits IFN-γ Secretion by Microglia to Negatively Regulate Anti-Tuberculosis Immunity in Mice. Microorganisms 2023; 11:2500. [PMID: 37894158 PMCID: PMC10608824 DOI: 10.3390/microorganisms11102500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/30/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Leucine-rich repeat-containing protein-25 (LRRC25) can degrade the ISG15 gene in virus-infected cells and prevent overactivation of the type Ⅰ IFN pathway. However, the role of LRRC25 in bacterial infection is still unclear. In this pursuit, the present study aimed to explore the regulatory role and mechanism of LRRC25 in microglia infected with Mycobacterium tuberculosis in a mouse model. METHODS Q-PCR, WB, and cell immunofluorescence were employed to observe the change in LRRC25 in BV2 cells infected by H37Rv. Additionally, siRNA was designed to target the LRRC25 to inhibit its expression in BV2 cells. Flow cytometry and laser confocal imaging were used to observe the infection of BV2 cells after LRRC25 silencing. Q-PCR and ELISA were used to determine the changes in IFN-γ and ISG15 in the culture supernatant of each group. RESULTS Following H37Rv infection, it was observed that the expression of LRRC25 was upregulated. Upon silencing LRRC25, the proportion of BV2 cells infected by H37Rv decreased significantly. ELISA analysis showed that IFN-γ and ISG15 levels in cell culture supernatant decreased after H37Rv infection, while they significantly increased after LRRC25 silencing. CONCLUSIONS This study provides evidence that LRRC25 is the key negative regulator of microglial anti-Mtb immunity. It exerts its function by degrading free ISG15 and inhibiting the secretion of IFN-γ, thereby improving the anti-Mtb immunity of BV2 cells.
Collapse
Affiliation(s)
- Gang Sheng
- Beijing Chest Hospital Affiliated to Capital Medical University, Beijing 100000, China; (G.S.); (H.C.); (W.W.); (N.T.); (D.L.)
| | - Hongqian Chu
- Beijing Chest Hospital Affiliated to Capital Medical University, Beijing 100000, China; (G.S.); (H.C.); (W.W.); (N.T.); (D.L.)
- Beijing Thoracic Tumor and Tuberculosis Institute, Beijing 100000, China;
| | - Huijuan Duan
- Beijing Thoracic Tumor and Tuberculosis Institute, Beijing 100000, China;
| | - Wenjing Wang
- Beijing Chest Hospital Affiliated to Capital Medical University, Beijing 100000, China; (G.S.); (H.C.); (W.W.); (N.T.); (D.L.)
| | - Na Tian
- Beijing Chest Hospital Affiliated to Capital Medical University, Beijing 100000, China; (G.S.); (H.C.); (W.W.); (N.T.); (D.L.)
| | - Dingyi Liu
- Beijing Chest Hospital Affiliated to Capital Medical University, Beijing 100000, China; (G.S.); (H.C.); (W.W.); (N.T.); (D.L.)
| | - Hong Sun
- Beijing Chest Hospital Affiliated to Capital Medical University, Beijing 100000, China; (G.S.); (H.C.); (W.W.); (N.T.); (D.L.)
- Beijing Thoracic Tumor and Tuberculosis Institute, Beijing 100000, China;
| | - Zhaogang Sun
- Beijing Chest Hospital Affiliated to Capital Medical University, Beijing 100000, China; (G.S.); (H.C.); (W.W.); (N.T.); (D.L.)
- Beijing Thoracic Tumor and Tuberculosis Institute, Beijing 100000, China;
| |
Collapse
|
11
|
Thuner J, Coutant F. IFN-γ: An overlooked cytokine in dermatomyositis with anti-MDA5 antibodies. Autoimmun Rev 2023; 22:103420. [PMID: 37625674 DOI: 10.1016/j.autrev.2023.103420] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
Dermatomyositis with anti-melanoma differentiation-associated gene 5 antibody (anti-MDA5 DM) is a rare autoimmune disease, often complicated by life-threatening, rapidly progressive interstitial lung disease. Additional manifestations of the disease include skin lesions, vascular abnormalities, joints and muscles pain. Despite its clinical significance, the pathogenesis of anti-MDA5 DM remains largely unknown. Currently, the disease is perceived as driven by type I interferon (IFN) whose expression is increased in most of the patients. Importantly, the regulation of IFN-γ is also altered in anti-MDA5 DM as evidenced by the presence of IFN-γ positive histiocytes in the lungs of patients, and the identification of autoantibodies that directly stimulate the production of IFN-γ by mononuclear cells. This review critically examines the pathogenesis of the disease, shedding light on recent findings that emphasize a potential role of IFN-γ. A novel conceptual framework is proposed, which integrates the molecular mechanisms altering IFN-γ regulation in anti-MDA5 DM with the known functional effects of IFN-γ on key tissues affected during the disease, such as the lungs, skin, and vessels. Understanding the precise role and relevance of IFN-γ in the pathogenesis of the disease will not only enhance the selection of available therapies for anti-MDA5 DM patients but also pave the way for the development of new therapeutic approaches targeting the altered molecular pathways.
Collapse
Affiliation(s)
- Jonathan Thuner
- Immunogenomics and Inflammation Research Team, University of Lyon, Edouard Herriot Hospital, Lyon, France; Internal medicine Department, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Frédéric Coutant
- Immunogenomics and Inflammation Research Team, University of Lyon, Edouard Herriot Hospital, Lyon, France; Immunology Department, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France.
| |
Collapse
|
12
|
Cui S, Zhang Z, Cheng C, Tang S, Zhai M, Li L, Wei F, Ding G. Small Extracellular Vesicles from Periodontal Ligament Stem Cells Primed by Lipopolysaccharide Regulate Macrophage M1 Polarization via miR-433-3p Targeting TLR2/TLR4/NF-κB. Inflammation 2023; 46:1849-1858. [PMID: 37351818 PMCID: PMC10567992 DOI: 10.1007/s10753-023-01845-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 06/24/2023]
Abstract
Lipopolysaccharide (LPS) is regarded as the main pathogenic factor of periodontitis. Mesenchymal stem cell-derived small extracellular vesicles (sEVs) play a key role in a variety of physiological and pathological processes. This study investigated the effects of sEVs derived from periodontal ligament stem cells (PDLSCs) pretreated with LPS on macrophage polarization and the underlying mechanisms. PDLSCs were treated with LPS (1 µg/mL) for 24 h, and sEVs were harvested by gradient centrifugation method. Macrophages were incubated with sEVs for 24 h, followed by examination of the expression profiles of inflammatory and anti-inflammatory cytokines, and polarization markers. Furthermore, microarray analysis, western blot test, and microRNA inhibitor transfection experiments were used to elucidate the molecular signaling pathway responsible for the process. The results showed that sEVs derived from LPS-preconditioning PDLSCs could significantly increase the expression of M1 markers and inflammatory cytokines, whereas decreased the expression of M2 markers and anti-inflammatory cytokines. Mechanistic analysis showed that TLR2/TLR4/NF-κB p65 pathway was involved in M1 polarization of macrophages, and microRNA-433-3p played a role, at least in part, in the course. Collectively, LPS could promote the macrophages into M1 status via TLR2/TLR4/NF-κB p65 signaling pathway partly by sEV-mediated microRNA-433-3p, which could be a potential therapeutic target for periodontitis.
Collapse
Affiliation(s)
- Shuyue Cui
- School of Stomatology, Weifang Medical University, Baotong West Street No. 7166, Weifang, Shandong, China
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, Jinan, Shandong, China
| | - Zijie Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, Jinan, Shandong, China
| | - Chen Cheng
- School of Stomatology, Weifang Medical University, Baotong West Street No. 7166, Weifang, Shandong, China
| | - Shuai Tang
- School of Stomatology, Weifang Medical University, Baotong West Street No. 7166, Weifang, Shandong, China
| | - Mingrui Zhai
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, Jinan, Shandong, China
| | - Lan Li
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, Jinan, Shandong, China
| | - Fulan Wei
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No.44-1 Wenhua Road West, Jinan, Shandong, China.
| | - Gang Ding
- School of Stomatology, Weifang Medical University, Baotong West Street No. 7166, Weifang, Shandong, China.
| |
Collapse
|
13
|
Zhang Y, Ma S, Li T, Tian Y, Zhou H, Wang H, Huang L. ILC1-derived IFN-γ regulates macrophage activation in colon cancer. Biol Direct 2023; 18:56. [PMID: 37679802 PMCID: PMC10486120 DOI: 10.1186/s13062-023-00401-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/26/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) are an important subset of innate immune cells in the tumor microenvironment, and they are pivotal regulators of tumor-promoting inflammation and tumor progression. Evidence has proven that TAM numbers are substantially increased in cancers, and most of these TAMs are polarized toward the alternatively activated M2 phenotype; Thus, these TAMs strongly promote the progression of cancer diseases. Type 1 innate lymphocytes (ILC1s) are present in high numbers in intestinal tissues and are characterized by the expression of the transcription factor T-bet and the secretion of interferon (IFN)-γ, which can promote macrophages to polarize toward the classically activated antitumor M1 phenotype. However, the relationship between these two cell subsets in colon cancer remains unclear. METHODS Flow cytometry was used to determine the percentages of M1-like macrophages, M2-like macrophages and ILC1s in colon cancer tissues and paracancerous healthy colon tissues in the AOM/DSS-induced mouse model of colon cancer. Furthermore, ILC1s were isolated and bone marrow-derived macrophages were generated to analyze the crosstalk that occurred between these cells when cocultured in vitro. Moreover, ILC1s were adoptively transferred or inhibited in vivo to explore the effects of ILC1s on tumor-infiltrating macrophages and tumor growth. RESULTS We found that the percentages of M1-like macrophages and ILC1s were decreased in colon cancer tissues, and these populations were positively correlated. ILC1s promoted the polarization of macrophages toward the classically activated M1-like phenotype in vitro, and this effect could be blocked by an anti-IFN-γ antibody. The in vivo results showed that the administration of the Group 1 innate lymphocyte-blocking anti-NK1.1 antibody decreased the number of M1-like macrophages in the tumor tissues of MC38 tumor-bearing mice and promoted tumor growth, and adoptive transfer of ILC1s inhibited tumors and increased the percentage of M1-like macrophages in MC38 tumor-bearing mice. CONCLUSIONS Our studies preliminarily prove for the first time that ILC1s promote the activation of M1-like macrophages by secreting IFN-γ and inhibit the progression of colon cancer, which may provide insight into immunotherapeutic approaches for colon cancer.
Collapse
Affiliation(s)
- Yandong Zhang
- Department of Rheumatology, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Shu Ma
- Department of Rheumatology, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Tie Li
- Department of Rheumatology, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Yu Tian
- Department of Laboratory Medicine, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, People's Republic of China
| | - Huangao Zhou
- Department of emergency medicine, Jiangyin People's Hospital, Wuxi, China.
| | - Hongsheng Wang
- Department of General Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou, China.
| | - Lan Huang
- Department of Laboratory Medicine, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, People's Republic of China.
| |
Collapse
|
14
|
Cortés H, Castillo-Ruiz M, Cañon-Jones H, Schlotterbeck T, San Martín R, Padilla L. In Vivo Efficacy of Purified Quillaja Saponin Extracts in Protecting against Piscirickettsia salmonis Infections in Atlantic Salmon ( Salmo salar). Animals (Basel) 2023; 13:2845. [PMID: 37760245 PMCID: PMC10525856 DOI: 10.3390/ani13182845] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/18/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Piscirickettsiosis, the main infectious disease affecting salmon farming in Chile, still has no efficient control measures. Piscirickettsia salmonis is a facultative intracellular bacterium that can survive and replicate within the host macrophages, evading the immune response. Triterpenic saponins obtained from the Quillaja saponaria tree have been widely studied, and have been shown to be immunomodulatory agents, suitable for feed and vaccine applications for veterinary and human uses. The impact of the oral administration of two extracts of Quillaja saponins on the infection of P. salmonis in Salmo salar and the corresponding gene expressions of immunomarkers were studied under three in vivo models. In the intraperitoneal challenge model, the group fed with Quillaja extracts showed lower mortality (29.1% treated vs. 37.5% control). Similar results were obtained in the cohabitation model trial (36.3% vs. 60.0%). In the commercial pilot trial, the results showed a significant reduction of 71.3% in mortality caused by P. salmonis (0.51% vs. 1.78%) and antibiotic use (reduction of 66.6% compared to untreated control). Also, Quillaja extracts significantly modulated the expression of IFN-II and CD8. These results represent evidence supporting the future use of purified Quillaja extracts as a natural non-pharmacological strategy for the prevention and control of P. salmonis infections in salmon.
Collapse
Affiliation(s)
- Hernán Cortés
- Desert King Chile, Viña del Mar 2420505, Chile; (T.S.); (L.P.)
| | - Mario Castillo-Ruiz
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago 8370134, Chile;
- Departamento de Ciencias Químicas y Biológicas, Facultad de Ciencias de la Salud, Universidad Bernardo O’Higgins, Santiago 8370854, Chile
| | - Hernán Cañon-Jones
- Núcleo de Investigación Aplicada en Ciencias Veterinarias y Agronómicas, Facultad de Medicina Veterinaria y Agronomía, Universidad de Las Américas, Santiago 7500975, Chile
| | | | - Ricardo San Martín
- Sutardja Center for Entrepreneurship and Technology, College of Engineering, University of California, Berkeley, CA 94720, USA;
| | - Leandro Padilla
- Desert King Chile, Viña del Mar 2420505, Chile; (T.S.); (L.P.)
| |
Collapse
|
15
|
Han J, Wu M, Liu Z. Dysregulation in IFN-γ signaling and response: the barricade to tumor immunotherapy. Front Immunol 2023; 14:1190333. [PMID: 37275859 PMCID: PMC10233742 DOI: 10.3389/fimmu.2023.1190333] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/14/2023] [Indexed: 06/07/2023] Open
Abstract
Interferon-gamma (IFN-γ) has been identified as a crucial factor in determining the responsiveness to immunotherapy. Produced primarily by natural killer (NK) and T cells, IFN-γ promotes activation, maturation, proliferation, cytokine expression, and effector function in immune cells, while simultaneously inducing antigen presentation, growth arrest, and apoptosis in tumor cells. However, tumor cells can hijack the IFN-γ signaling pathway to mount IFN-γ resistance: rather than increasing antigenicity and succumbing to death, tumor cells acquire stemness characteristics and express immunosuppressive molecules to defend against antitumor immunity. In this review, we summarize the potential mechanisms of IFN-γ resistance occurring at two critical stages: disrupted signal transduction along the IFNG/IFNGR/JAK/STAT pathway, or preferential expression of specific interferon-stimulated genes (ISGs). Elucidating the molecular mechanisms through which tumor cells develop IFN-γ resistance help identify promising therapeutic targets to improve immunotherapy, with broad application value in conjugation with targeted, antibody or cellular therapies.
Collapse
Affiliation(s)
- Jiashu Han
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of General Surgery, Peking Union Medical College Hospital (CAMS), Beijing, China
| | - Mengwei Wu
- Department of General Surgery, Peking Union Medical College Hospital (CAMS), Beijing, China
| | - Ziwen Liu
- Department of General Surgery, Peking Union Medical College Hospital (CAMS), Beijing, China
| |
Collapse
|
16
|
Han J, Dong L, Wu M, Ma F. Dynamic polarization of tumor-associated macrophages and their interaction with intratumoral T cells in an inflamed tumor microenvironment: from mechanistic insights to therapeutic opportunities. Front Immunol 2023; 14:1160340. [PMID: 37251409 PMCID: PMC10219223 DOI: 10.3389/fimmu.2023.1160340] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/20/2023] [Indexed: 05/31/2023] Open
Abstract
Immunotherapy has brought a paradigm shift in the treatment of tumors in recent decades. However, a significant proportion of patients remain unresponsive, largely due to the immunosuppressive tumor microenvironment (TME). Tumor-associated macrophages (TAMs) play crucial roles in shaping the TME by exhibiting dual identities as both mediators and responders of inflammation. TAMs closely interact with intratumoral T cells, regulating their infiltration, activation, expansion, effector function, and exhaustion through multiple secretory and surface factors. Nevertheless, the heterogeneous and plastic nature of TAMs renders the targeting of any of these factors alone inadequate and poses significant challenges for mechanistic studies and clinical translation of corresponding therapies. In this review, we present a comprehensive summary of the mechanisms by which TAMs dynamically polarize to influence intratumoral T cells, with a focus on their interaction with other TME cells and metabolic competition. For each mechanism, we also discuss relevant therapeutic opportunities, including non-specific and targeted approaches in combination with checkpoint inhibitors and cellular therapies. Our ultimate goal is to develop macrophage-centered therapies that can fine-tune tumor inflammation and empower immunotherapy.
Collapse
Affiliation(s)
- Jiashu Han
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing, China
| | - Luochu Dong
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing, China
| | - Mengwei Wu
- Department of General Surgery, Peking Union Medical College Hospital (CAMS), Beijing, China
| | - Fei Ma
- Center for National Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
17
|
Xiao LP, Zhou LN, Chen JJ, Zhang Y, Tang XM, Zhou J. [A preliminary study on the role of V-domain Ig suppressor of T cell activation in juvenile idiopathic arthritis]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:272-277. [PMID: 36946162 PMCID: PMC10032065 DOI: 10.7499/j.issn.1008-8830.2211105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
OBJECTIVES To study the expression of V-domain Ig suppressor of T cell activation (VISTA) in peripheral blood of children with juvenile idiopathic arthritis (JIA) and its role in the pathogenesis of JIA. METHODS In this prospective study, peripheral blood was collected from 47 children with different subtypes of JIA and 10 healthy children. Flow cytometry was used to measure the expression levels of VISTA, interferon-γ (IFN-γ), and tumor necrosis factor-α (TNF-α) on CD14+ mononuclear cells, CD4+ T lymphocytes, and CD8+ T lymphocytes. RESULTS The children with JIA had a significantly lower expression level of VISTA than the healthy children (P<0.05). There was a significant difference in the expression of VISTA between the children with different subtypes of JIA, with the lowest expression level in those with systemic JIA (P<0.05). There was also a significant difference in the expression of VISTA between different immune cells, with a significantly higher expression level on the surface of monocytes (P<0.05). Correlation analysis showed that VISTA was negatively correlated with the expression of IFN-γ and TNF-α on CD4+ T cells (r=-0.436 and -0.382 respectively, P<0.05), CD8+ T cells (r=-0.348 and -0.487 respectively, P<0.05), and CD14+ mononuclear cells (r=-0.582 and -0.603 respectively, P<0.05). CONCLUSIONS The insufficient expression of VISTA may be associated with the pathogenesis of JIA, and enhancing the immunomodulatory effect of VISTA might be one option for the treatment of JIA in the future.
Collapse
Affiliation(s)
- Li-Ping Xiao
- Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University/National Clinical Research Center for Child Health and Disorders/China International Science and Technology Cooperation Base of Child Development and Critical Disorders/Chongqing Key Laboratory of Child Infection and Immunity/Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
| | - Li-Na Zhou
- Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University/National Clinical Research Center for Child Health and Disorders/China International Science and Technology Cooperation Base of Child Development and Critical Disorders/Chongqing Key Laboratory of Child Infection and Immunity/Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
| | - Jun-Jie Chen
- Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University/National Clinical Research Center for Child Health and Disorders/China International Science and Technology Cooperation Base of Child Development and Critical Disorders/Chongqing Key Laboratory of Child Infection and Immunity/Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
| | - Yan Zhang
- Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University/National Clinical Research Center for Child Health and Disorders/China International Science and Technology Cooperation Base of Child Development and Critical Disorders/Chongqing Key Laboratory of Child Infection and Immunity/Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
| | - Xue-Mei Tang
- Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University/National Clinical Research Center for Child Health and Disorders/China International Science and Technology Cooperation Base of Child Development and Critical Disorders/Chongqing Key Laboratory of Child Infection and Immunity/Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
| | - Juan Zhou
- Department of Rheumatology and Immunology, Children's Hospital of Chongqing Medical University/National Clinical Research Center for Child Health and Disorders/China International Science and Technology Cooperation Base of Child Development and Critical Disorders/Chongqing Key Laboratory of Child Infection and Immunity/Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing 400014, China
| |
Collapse
|
18
|
Umitaibatin R, Harisna AH, Jauhar MM, Syaifie PH, Arda AG, Nugroho DW, Ramadhan D, Mardliyati E, Shalannanda W, Anshori I. Immunoinformatics Study: Multi-Epitope Based Vaccine Design from SARS-CoV-2 Spike Glycoprotein. Vaccines (Basel) 2023; 11:vaccines11020399. [PMID: 36851275 PMCID: PMC9964839 DOI: 10.3390/vaccines11020399] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
The coronavirus disease 2019 outbreak has become a huge challenge in the human sector for the past two years. The coronavirus is capable of mutating at a higher rate than other viruses. Thus, an approach for creating an effective vaccine is still needed to induce antibodies against multiple variants with lower side effects. Currently, there is a lack of research on designing a multiepitope of the COVID-19 spike protein for the Indonesian population with comprehensive immunoinformatic analysis. Therefore, this study aimed to design a multiepitope-based vaccine for the Indonesian population using an immunoinformatic approach. This study was conducted using the SARS-CoV-2 spike glycoprotein sequences from Indonesia that were retrieved from the GISAID database. Three SARS-CoV-2 sequences, with IDs of EIJK-61453, UGM0002, and B.1.1.7 were selected. The CD8+ cytotoxic T-cell lymphocyte (CTL) epitope, CD4+ helper T lymphocyte (HTL) epitope, B-cell epitope, and IFN-γ production were predicted. After modeling the vaccines, molecular docking, molecular dynamics, in silico immune simulations, and plasmid vector design were performed. The designed vaccine is antigenic, non-allergenic, non-toxic, capable of inducing IFN-γ with a population reach of 86.29% in Indonesia, and has good stability during molecular dynamics and immune simulation. Hence, this vaccine model is recommended to be investigated for further study.
Collapse
Affiliation(s)
- Ramadhita Umitaibatin
- Lab-on-Chip Group, Department of Biomedical Engineering, School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung 40132, Indonesia
| | - Azza Hanif Harisna
- Nano Center Indonesia, Jl. Raya Puspiptek, South Tangerang 15314, Indonesia
| | | | - Putri Hawa Syaifie
- Nano Center Indonesia, Jl. Raya Puspiptek, South Tangerang 15314, Indonesia
| | | | - Dwi Wahyu Nugroho
- Nano Center Indonesia, Jl. Raya Puspiptek, South Tangerang 15314, Indonesia
| | - Donny Ramadhan
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), Cibinong 16911, Indonesia
| | - Etik Mardliyati
- Research Center for Vaccine and Drug, National Research and Innovation Agency (BRIN), Cibinong 16911, Indonesia
| | - Wervyan Shalannanda
- Department of Telecommunication Engineering, School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung 40132, Indonesia
| | - Isa Anshori
- Lab-on-Chip Group, Department of Biomedical Engineering, School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung 40132, Indonesia
- Correspondence:
| |
Collapse
|
19
|
Potential Pathophysiological Mechanisms Underlying Multiple Organ Dysfunction in Cytokine Release Syndrome. Mediators Inflamm 2022; 2022:7137900. [PMID: 35431655 PMCID: PMC9007670 DOI: 10.1155/2022/7137900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/16/2022] [Indexed: 12/15/2022] Open
Abstract
In recent decades, many serious respiratory infections have broken out all over the world, including SARS-CoV, MERS, and COVID-19. They are characterized by strong infectivity, rapid disease progression, high mortality, and poor prognosis. Excessive immune system activation results in cytokine hypersecretion, which is an important reason for the aggravation of symptoms, and can spread throughout the body leading to systemic multiple organ dysfunction, namely, cytokine release syndrome (CRS). Although many diseases related to CRS have been identified, the mechanism of CRS is rarely mentioned clearly. This review is intended to clarify the pathogenetic mechanism of CRS in the deterioration of related diseases, describe the important signaling pathways and clinical pathophysiological characteristics of CRS, and provide ideas for further research and development of specific drugs for corresponding targets to treat CRS.
Collapse
|
20
|
Szebeni J, Storm G, Ljubimova JY, Castells M, Phillips EJ, Turjeman K, Barenholz Y, Crommelin DJA, Dobrovolskaia MA. Applying lessons learned from nanomedicines to understand rare hypersensitivity reactions to mRNA-based SARS-CoV-2 vaccines. NATURE NANOTECHNOLOGY 2022; 17:337-346. [PMID: 35393599 DOI: 10.1038/s41565-022-01071-x] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 01/04/2022] [Indexed: 05/24/2023]
Abstract
After over a billion of vaccinations with messenger RNA-lipid nanoparticle (mRNA-LNP) based SARS-CoV-2 vaccines, anaphylaxis and other manifestations of hypersensitivity can be considered as very rare adverse events. Although current recommendations include avoiding a second dose in those with first-dose anaphylaxis, the underlying mechanisms are unknown; therefore, the risk of a future reaction cannot be predicted. Given how important new mRNA constructs will be to address the emergence of new viral variants and viruses, there is an urgent need for clinical approaches that would allow a safe repeated immunization of high-risk individuals and for reliable predictive tools of adverse reactions to mRNA vaccines. In many aspects, anaphylaxis symptoms experienced by the affected vaccine recipients resemble those of infusion reactions to nanomedicines. Here we share lessons learned over a decade of nanomedicine research and discuss the current knowledge about several factors that individually or collectively contribute to infusion reactions to nanomedicines. We aim to use this knowledge to inform the SARS-CoV-2 lipid-nanoparticle-based mRNA vaccine field.
Collapse
Affiliation(s)
- Janos Szebeni
- Nanomedicine Research and Education Center, Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
- SeroScience LCC, Budapest, Hungary
- Department of Nanobiotechnology and Regenerative Medicine, Faculty of Health, Miskolc University, Miskolc, Hungary
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, the Netherlands
- Department of Biomaterials Science and Technology, University of Twente, Enschede, the Netherlands
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Mariana Castells
- Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Elizabeth J Phillips
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Keren Turjeman
- Laboratory of Membrane and Liposome Research, IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Yechezkel Barenholz
- Laboratory of Membrane and Liposome Research, IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Daan J A Crommelin
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Marina A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD, USA.
| |
Collapse
|
21
|
Pro- and Anti-Inflammatory Cytokines in the Context of NK Cell-Trophoblast Interactions. Int J Mol Sci 2022; 23:ijms23042387. [PMID: 35216502 PMCID: PMC8878424 DOI: 10.3390/ijms23042387] [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: 01/26/2022] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 02/04/2023] Open
Abstract
During pregnancy, uterine NK cells interact with trophoblast cells. In addition to contact interactions, uterine NK cells are influenced by cytokines, which are secreted by the cells of the decidua microenvironment. Cytokines can affect the phenotypic characteristics of NK cells and change their functional activity. An imbalance of pro- and anti-inflammatory signals can lead to the development of reproductive pathology. The aim of this study was to assess the effects of cytokines on NK cells in the presence of trophoblast cells in an in vitro model. We used TNFα, IFNγ, TGFβ and IL-10; the NK-92 cell line; and peripheral blood NK cells (pNKs) from healthy, non-pregnant women. For trophoblast cells, the JEG-3 cell line was used. In the monoculture of NK-92 cells, TNFα caused a decrease in CD56 expression. In the coculture of NK cells with JEG-3 cells, TNFα increased the expression of NKG2C and NKG2A by NK-92 cells. Under the influence of TGFβ, the expression of CD56 increased and the expression of NKp30 decreased in the monoculture. After the preliminary cultivation of NK-92 cells in the presence of TGFβ, their cytotoxicity increased. In the case of adding TGFβ to the PBMC culture, as well as coculturing PBMCs and JEG-3 cells, the expression of CD56 and NKp44 by pNK cells was reduced. The differences in the effects of TGFβ in the model using NK-92 cells and pNK cells may be associated with the possible influence of monocytes or other lymphoid cells from the mononuclear fraction.
Collapse
|
22
|
Bergeron HC, Tripp RA. Immunopathology of RSV: An Updated Review. Viruses 2021; 13:2478. [PMID: 34960746 PMCID: PMC8703574 DOI: 10.3390/v13122478] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022] Open
Abstract
RSV is a leading cause of respiratory tract disease in infants and the elderly. RSV has limited therapeutic interventions and no FDA-approved vaccine. Gaps in our understanding of virus-host interactions and immunity contribute to the lack of biological countermeasures. This review updates the current understanding of RSV immunity and immunopathology with a focus on interferon responses, animal modeling, and correlates of protection.
Collapse
Affiliation(s)
| | - Ralph A. Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
23
|
Ruan F, Wu L, Yin H, Fang L, Tang C, Huang S, Fang L, Zuo Z, He C, Huang J. Long-term exposure to environmental level of phenanthrene causes adaptive immune response and fibrosis in mouse kidneys. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 283:117028. [PMID: 33892371 DOI: 10.1016/j.envpol.2021.117028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 03/10/2021] [Accepted: 03/24/2021] [Indexed: 06/12/2023]
Abstract
As ubiquitous, persistent organic pollutants, polycyclic aromatic hydrocarbons (PAHs) have adverse impacts on human health. Phenanthrene (Phe) is one of the most abundant PAHs in the environment. However, the long-term effects of exposure to environmental level of Phe on the kidneys and the potential mechanisms are unclear. T helper (Th) cells, a subtype of CD4+ T cells that play a central role in the renal immune microenvironment. In this study, male mice were chronically exposed to 5, 50, and 500 ng/kg bw Phe every other day for total 210 days. Those results indicated that environmental Phe exposure caused kidney hypertrophy, injury and fibrosis in the mice. Chronic, long-term environmental level of Phe exposure did not significantly alter the innate immune response but induced adaptive immune response changes (Th1/Th2 related cytokines release), causing a type 1 immune response in the 5 ng/kg bw Phe group and a type 2 immune response in the high dose groups (50 and 500 ng/kg bw). This study provides novel insights into the roles of adaptive immune response in long-term PAH exposure-induced chronic kidney injury and fibrosis, which is beneficial for further understanding the potential health hazards of PAHs and providing new avenues for immune intervention strategies to alleviate PAHs toxicity.
Collapse
Affiliation(s)
- Fengkai Ruan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The 5th Hospital of Xiamen, Xiang'an Branch of the First Affiliated Hospital, Xiamen University, Xiamen, Fujian, 361102, China
| | - Lifang Wu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 S. Chongqing Road, Shanghai, 200025, China
| | - Hanying Yin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The 5th Hospital of Xiamen, Xiang'an Branch of the First Affiliated Hospital, Xiamen University, Xiamen, Fujian, 361102, China
| | - Lu Fang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The 5th Hospital of Xiamen, Xiang'an Branch of the First Affiliated Hospital, Xiamen University, Xiamen, Fujian, 361102, China
| | - Chen Tang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The 5th Hospital of Xiamen, Xiang'an Branch of the First Affiliated Hospital, Xiamen University, Xiamen, Fujian, 361102, China
| | - Siyang Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The 5th Hospital of Xiamen, Xiang'an Branch of the First Affiliated Hospital, Xiamen University, Xiamen, Fujian, 361102, China
| | - Longxiang Fang
- Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The 5th Hospital of Xiamen, Xiang'an Branch of the First Affiliated Hospital, Xiamen University, Xiamen, Fujian, 361102, China
| | - Chengyong He
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The 5th Hospital of Xiamen, Xiang'an Branch of the First Affiliated Hospital, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jiyi Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The 5th Hospital of Xiamen, Xiang'an Branch of the First Affiliated Hospital, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
24
|
From Coxiella burnetii Infection to Pregnancy Complications: Key Role of the Immune Response of Placental Cells. Pathogens 2021; 10:pathogens10050627. [PMID: 34069587 PMCID: PMC8160966 DOI: 10.3390/pathogens10050627] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 01/17/2023] Open
Abstract
The infection of pregnant animals and women by Coxiella burnetii, an intracellular bacterium, compromises both maternal health and foetal development. The placenta is targeted by C. burnetii, as demonstrated by bacteriological and histological evidence. It now appears that placental strains of C. burnetii are highly virulent compared to reference strains and that placental injury involves different types of placental cells. Trophoblasts, the major placental cells, are largely infected by C. burnetii and may represent a replicating niche for the bacteria. The placenta also contains numerous immune cells, including macrophages, dendritic cells, and mast cells. Placental macrophages are infected and activated by C. burnetii in an unusual way of M1 polarisation associated with bacterial elimination. Placental mast cells eliminate bacteria through a mechanism including the release of extracellular actin filaments and antimicrobial peptides. In contrast, C. burnetii impairs the maturation of decidual dendritic cells, favouring bacterial pathogenicity. Our aim is to review C. burnetii infections of human placentas, paying special attention to both the action and function of the different cell types, immune cells, and trophoblasts targeted by C. burnetii in relation to foetal injury.
Collapse
|
25
|
Zhang C, Ju J, Wu X, Yang J, Yang Q, Liu C, Chen L, Sun X. Tripterygium wilfordii Polyglycoside Ameliorated TNBS-Induced Colitis in Rats via Regulating Th17/Treg Balance in Intestinal Mucosa. J Inflamm Res 2021; 14:1243-1255. [PMID: 33833546 PMCID: PMC8021269 DOI: 10.2147/jir.s293961] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/10/2021] [Indexed: 12/13/2022] Open
Abstract
Purpose To investigate the therapeutic effect of Tripterygium wilfordii polyglycoside (TWP), a derivative from a Chinese traditional herb, on 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis, in a model for inflammatory bowel disease (IBD) in rats. Methods TWP was administrated to Wistar rats during TNBS-induced colitis to determine its therapeutic effect on active inflammation using the Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR), flow cytometry, and Western blotting. Peripheral blood CD4+ T-cells were isolated from patients with ulcerative colitis (UC) and incubated with TWP to verify its immune regulation mechanism by qRT-PCR and flow cytometry. Results Intragastric administration of TWP attenuated the severity of intestinal inflammation in TNBS-induced rat colitis, characterized by decreased DAI, histopathological scores, and expression of IL-6, TNFα, IFNγ, and IL-17A in intestinal mucosa. Furthermore, TWP reduced IL-17A+CD4+ T-cells, while enhanced Foxp3+CD25+CD4+ T-cells in peripheral blood, mesenteric lymph nodes (MLN), and spleen in rat colitis. Downstream signaling including ROR-γt, STAT3, and HIF1α expression in intestinal mucosa were suppressed by TWP. In addition, incubation with TWP suppressed IL-17A+CD4+ T-cell differentiation, while it promoted Foxp3+CD25+CD4+ T-cell differentiation in CD4+ T-cells isolated from UC patients. Conclusion TWP successfully ameliorated experimental rat colitis via regulating innate immune responses as well as Th17/Treg balance in intestinal mucosa, peripheral blood, MLN, and spleen. Moreover, the differentiation of peripheral blood CD4+ T-cell isolated from patients with UC was modulated by TWP. TWP may act as an optional complementary and alternative medicine for IBD.
Collapse
Affiliation(s)
- Cui Zhang
- Gastroenterology Department, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Jingyi Ju
- Gastroenterology Department, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Xiaohan Wu
- Gastroenterology Department, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Jiaolan Yang
- Gastroenterology Department, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Qinglu Yang
- Gastroenterology Department, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Changqin Liu
- Gastroenterology Department, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Liang Chen
- Gastroenterology Department, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Xiaomin Sun
- Gastroenterology Department, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, People's Republic of China.,Gastroenterology Department, The Shanghai Tenth People's Hospital, Chongming Branch, Shanghai, People's Republic of China
| |
Collapse
|
26
|
PlGF Immunological Impact during Pregnancy. Int J Mol Sci 2020; 21:ijms21228714. [PMID: 33218096 PMCID: PMC7698813 DOI: 10.3390/ijms21228714] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022] Open
Abstract
During pregnancy, the mother’s immune system has to tolerate the persistence of paternal alloantigens without affecting the anti-infectious immune response. Consequently, several mechanisms aimed at preventing allograft rejection, occur during a pregnancy. In fact, the early stages of pregnancy are characterized by the correct balance between inflammation and immune tolerance, in which proinflammatory cytokines contribute to both the remodeling of tissues and to neo-angiogenesis, thus, favoring the correct embryo implantation. In addition to the creation of a microenvironment able to support both immunological privilege and angiogenesis, the trophoblast invades normal tissues by sharing the same behavior of invasive tumors. Next, the activation of an immunosuppressive phase, characterized by an increase in the number of regulatory T (Treg) cells prevents excessive inflammation and avoids fetal immuno-mediated rejection. When these changes do not occur or occur incompletely, early pregnancy failure follows. All these events are characterized by an increase in different growth factors and cytokines, among which one of the most important is the angiogenic growth factor, namely placental growth factor (PlGF). PlGF is initially isolated from the human placenta. It is upregulated during both pregnancy and inflammation. In this review, we summarize current knowledge on the immunomodulatory effects of PlGF during pregnancy, warranting that both innate and adaptive immune cells properly support the early events of implantation and placental development. Furthermore, we highlight how an alteration of the immune response, associated with PlGF imbalance, can induce a hypertensive state and lead to the pre-eclampsia (PE).
Collapse
|