1
|
Valério-Bolas A, Meunier M, Rodrigues A, Palma-Marques J, Ferreira R, Cardoso I, Lobo L, Monteiro M, Nunes T, Armada A, Antunes WT, Alexandre-Pires G, da Fonseca IP, Santos-Gomes G. Unveiling the Interplay Between Dendritic Cells and Natural Killer Cells as Key Players in Leishmania Infection. J Immunol Res 2025; 2025:3176927. [PMID: 39963187 PMCID: PMC11832263 DOI: 10.1155/jimr/3176927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 01/02/2025] [Indexed: 02/20/2025] Open
Abstract
Leishmaniasis is a group of parasitic diseases whose etiological agent is the protozoa Leishmania. These diseases afflict impoverished populations in tropical and subtropical regions and affect wild and domestic animals. Canine leishmaniasis is a global disease mostly caused by L. infantum. Dogs are recognized as a good reservoir since harbor the infection long before developing the disease, facilitating parasite transmission. Furthermore, there is growing evidence that dogs may also be the reservoir of the American Leishmania spp. as L. amazonensis. The innate immune response is the first defense line against pathogens, which includes natural killer (NK) and dendritic cells (DCs). By recognizing and ultimately destroying infected cells, and by secreting immune mediators that favor inflammatory microenvironments, NK cells take the lead in the infectious process. When interacting with Leishmania parasites, DCs become activated and play a key role in driving the host immune response. While activated DCs can modulate NK cell activity, Leishmania parasites can directly activate NK cells by interacting with innate immune receptors. Once activated, NK cells can engage in a bidirectional interplay with DCs. However, the complexity of these interactions during Leishmania infection makes it challenging to fully understand the underlying processes. To further explore this, the present study investigated the dynamic interplay established between monocyte-derived DCs (moDCs) and putative NK (pNK) cells of dogs during Leishmania infection. Findings indicate that the crosstalk between moDCs exposed to L. infantum or L. amazonensis and pNK cells enhances chemokine upregulation, potentially attracting other leukocytes to the site of infection. pNK cells activated by L. infantum infected DCs upregulate IL-10, which can lead to a regulatory immune response while moDCs exposed to L. amazonensis induced pNK cells to overexpress IFN-γ and IL-13, favoring a mix of pro- and anti-inflammatory response. In addition, parasite-derived extracellular vesicles (EVs) can modulate the host immune response by stimulating the upregulation of anti-inflammatory cytokines and perforin release, which may impact infection outcomes. Thus, Leishmania and parasitic EVs can influence the bidirectional interplay between canine NK cells and DCs.
Collapse
Affiliation(s)
- Ana Valério-Bolas
- Unit for Teaching and Research in Medical Parasitology, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Lisbon 1349-008, Portugal
| | - Mafalda Meunier
- Unit for Teaching and Research in Medical Parasitology, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Lisbon 1349-008, Portugal
| | - Armanda Rodrigues
- Unit for Teaching and Research in Medical Parasitology, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Lisbon 1349-008, Portugal
| | - Joana Palma-Marques
- Unit for Teaching and Research in Medical Parasitology, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Lisbon 1349-008, Portugal
| | - Rui Ferreira
- BSA, Banco de Sangue Animal, Porto 4100-462, Portugal
| | - Inês Cardoso
- BSA, Banco de Sangue Animal, Porto 4100-462, Portugal
| | - Lis Lobo
- Unit for Teaching and Research in Medical Parasitology, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Lisbon 1349-008, Portugal
| | - Marta Monteiro
- Unit for Teaching and Research in Medical Parasitology, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Lisbon 1349-008, Portugal
- Faculty of Veterinary Medicine, Centre for Interdisciplinary Research in Animal Health, CIISA, University of Lisbon, Av. Universidade Técnica, Lisbon 1300-477, Portugal
| | - Telmo Nunes
- Microscopy Center, Faculty of Sciences of the University of Lisbon-FCUL—BioISI Ce3CE, Lisboa, Portugal
| | - Ana Armada
- Unit for Teaching and Research in Medical Parasitology, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Lisbon 1349-008, Portugal
| | - Wilson T. Antunes
- Instituto Universitário Militar (IUM), Centro de Investigação, Desenvolvimento e Inovação da Academia Militar (CINAMIL), Unidade Militar Laboratorial de Defesa Biológica e Química (UMLDBQ), Lisboa 1849-012, Portugal
| | - Graça Alexandre-Pires
- Faculty of Veterinary Medicine, Centre for Interdisciplinary Research in Animal Health, CIISA, University of Lisbon, Av. Universidade Técnica, Lisbon 1300-477, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Isabel Pereira da Fonseca
- Faculty of Veterinary Medicine, Centre for Interdisciplinary Research in Animal Health, CIISA, University of Lisbon, Av. Universidade Técnica, Lisbon 1300-477, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Gabriela Santos-Gomes
- Unit for Teaching and Research in Medical Parasitology, Global Health and Tropical Medicine, GHTM, Associate Laboratory in Translation and Innovation Towards Global Health, LA-REAL, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Lisbon 1349-008, Portugal
| |
Collapse
|
2
|
Maia AR, Gonzalez L, Bounab B, Grassi L, Mousset C, Fromont-Hankard G, Cezard A, Hiemstra P, Baranek T, Paget C, Crabbé A, Si-Tahar M. Intranasal exposure to commensal bacterium Rothia mucilaginosa protects against influenza A virus infection. Antiviral Res 2025; 234:106076. [PMID: 39755332 DOI: 10.1016/j.antiviral.2025.106076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/28/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
The respiratory tract hosts a diverse microbial community whose composition varies with anatomical location and throughout life. Rothia mucilaginosa, a common commensal of the upper respiratory tract and oral cavity, has recently been recognized for its ability to inhibit bacteria-triggered pro-inflammatory responses. However, its role in modulating the immune response to viral infections such as influenza A virus (IAV) pneumonia, remains unknown. Here, we demonstrate that R. mucilaginosa enhances protection against IAV, promoting viral clearance, reducing inflammation, preserving bronchial and alveolar structures, and improving survival in a mouse model of influenza pneumonia. The enhanced viral clearance observed in R. mucilaginosa-treated mice is associated with the recruitment of innate immune cells to the lungs, including PD-L1-expressing neutrophils, alongside the production of the anti-inflammatory cytokine IL-10, both of which are known to play regulatory roles in the context of IAV infection. Together, these findings highlight R. mucilaginosa-mediated innate immune priming as a key protective mechanism in the respiratory tract against IAV infection.
Collapse
Affiliation(s)
- Ana Raquel Maia
- INSERM, Research Center for Respiratory Diseases, UMR 1100, University of Tours, France
| | - Loïc Gonzalez
- INSERM, Research Center for Respiratory Diseases, UMR 1100, University of Tours, France
| | - Badreddine Bounab
- INSERM, Research Center for Respiratory Diseases, UMR 1100, University of Tours, France
| | - Lucia Grassi
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Coralie Mousset
- Department of Pathology, CHU of Tours, University of Tours, Tours, France
| | | | - Adeline Cezard
- INSERM, Research Center for Respiratory Diseases, UMR 1100, University of Tours, France
| | - Pieter Hiemstra
- Department of Pulmonology, Leiden University Medical Centre, Leiden, Netherlands
| | - Thomas Baranek
- INSERM, Research Center for Respiratory Diseases, UMR 1100, University of Tours, France
| | - Christophe Paget
- INSERM, Research Center for Respiratory Diseases, UMR 1100, University of Tours, France
| | - Aurélie Crabbé
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium.
| | - Mustapha Si-Tahar
- INSERM, Research Center for Respiratory Diseases, UMR 1100, University of Tours, France.
| |
Collapse
|
3
|
Lodi L, Voarino M, Stocco S, Ricci S, Azzari C, Galli L, Chiappini E. Immune response to viscerotropic Leishmania: a comprehensive review. Front Immunol 2024; 15:1402539. [PMID: 39359727 PMCID: PMC11445144 DOI: 10.3389/fimmu.2024.1402539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 08/15/2024] [Indexed: 10/04/2024] Open
Abstract
L. donovani and L. infantum infections are associated with a broad clinical spectrum, ranging from asymptomatic cases to visceral leishmaniasis (VL) with high mortality rates. Clinical manifestations such as post-kala-azar dermal leishmaniasis (PKDL) and visceral leishmaniasis-associated hemophagocytic lymphohistiocytosis-mimic (VL-associated HLH-mimic) further contribute to the diversity of clinical manifestations. These clinical variations are intricately influenced by the complex interplay between the host's immune response and the parasite's escape mechanisms. This narrative review aims to elucidate the underlying immunological mechanisms associated with each clinical manifestation, drawing from published literature within the last 5 years. Specific attention is directed toward viscerotropic Leishmania sinfection in patients with inborn errors of immunity and acquired immunodeficiencies. In VL, parasites exploit various immune evasion mechanisms, including immune checkpoints, leading to a predominantly anti-inflammatory environment that favors parasite survival. Conversely, nearly 70% of individuals are capable of mounting an effective pro-inflammatory immune response, forming granulomas that contain the parasites. Despite this, some patients may experience reactivation of the disease upon immunosuppression, challenging current understandings of parasite eradication. Individuals living with HIV and those with inborn errors of immunity present a more severe course of infection, often with higher relapse rates. Therefore, it is crucial to exclude both primary and acquired immune deficiencies in patients presenting disease relapse and VL-associated HLH-mimic. The distinction between VL and HLH can be challenging due to clinical similarities, suggesting that the nosological entity known as VL-associated HLH may represent a severe presentation of symptomatic VL and it should be considered more accurate referring to this condition as VL-associated HLH-mimic. Consequently, excluding VL in patients presenting with HLH is essential, as appropriate antimicrobial therapy can reverse immune dysregulation. A comprehensive understanding of the immune-host interaction underlying Leishmania infection is crucial for formulating effective treatment and preventive strategies to mitigate the disease burden.
Collapse
Affiliation(s)
- Lorenzo Lodi
- Department of Health Sciences, University of Florence, Florence, Italy
- Immunology Unit, Department of Pediatrics, Meyer Children’s Hospital IRCCS, Florence, Italy
| | - Marta Voarino
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Silvia Stocco
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Silvia Ricci
- Department of Health Sciences, University of Florence, Florence, Italy
- Immunology Unit, Department of Pediatrics, Meyer Children’s Hospital IRCCS, Florence, Italy
| | - Chiara Azzari
- Department of Health Sciences, University of Florence, Florence, Italy
- Immunology Unit, Department of Pediatrics, Meyer Children’s Hospital IRCCS, Florence, Italy
| | - Luisa Galli
- Department of Health Sciences, University of Florence, Florence, Italy
- Infectious Diseases Unit, Department of Pediatrics, Meyer Children’s Hospital IRCCS, Florence, Italy
| | - Elena Chiappini
- Department of Health Sciences, University of Florence, Florence, Italy
- Infectious Diseases Unit, Department of Pediatrics, Meyer Children’s Hospital IRCCS, Florence, Italy
| |
Collapse
|
4
|
Pollenus E, Possemiers H, Knoops S, Prenen F, Vandermosten L, Pham TT, Buysrogge L, Matthys P, Van den Steen PE. NK cells contribute to the resolution of experimental malaria-associated acute respiratory distress syndrome after antimalarial treatment. Front Immunol 2024; 15:1433904. [PMID: 39355242 PMCID: PMC11442241 DOI: 10.3389/fimmu.2024.1433904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/27/2024] [Indexed: 10/03/2024] Open
Abstract
In both humans and mice, natural killer (NK) cells are important lymphocytes of the innate immune system. They are often considered pro-inflammatory effector cells but may also have a regulatory or pro-resolving function by switching their cytokine profile towards the production of anti-inflammatory cytokines, including interleukin-10 (IL-10) and transforming growth factor-β, and by killing pro-inflammatory immune cells. Here, the role of NK cells in the resolution of malaria lung pathology was studied. Malaria complications, such as malaria-associated acute respiratory distress syndrome (MA-ARDS), are often lethal despite the rapid and efficient killing of Plasmodium parasites with antimalarial drugs. Hence, studying the resolution and healing mechanisms involved in the recovery from these complications could be useful to develop adjunctive treatments. Treatment of Plasmodium berghei NK65-infected C57BL/6 mice with a combination of artesunate and chloroquine starting at the appearance of symptoms was used as a model to study the resolution of MA-ARDS. The role of NK cells was studied using anti-NK1.1 depletion antibodies and NK cell-deficient mice. Using both methods, NK cells were found to be dispensable in the development of MA-ARDS, as shown previously. In contrast, NK cells were crucial in the initiation of resolution upon antimalarial treatment, as survival was significantly decreased in the absence of NK cells. Considerably increased IL-10 expression by NK cells suggested an anti-inflammatory and pro-resolving phenotype. Despite the increase in Il10 expression in the NK cells, inhibition of the IL-10/IL-10R axis using anti-IL10R antibodies had no effect on the resolution for MA-ARDS, suggesting that the pro-resolving effect of NK cells cannot solely be attributed to their IL-10 production. In conclusion, NK cells contribute to the resolution of experimental MA-ARDS.
Collapse
Affiliation(s)
- Emilie Pollenus
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Hendrik Possemiers
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Sofie Knoops
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Fran Prenen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Leen Vandermosten
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Thao-Thy Pham
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
- Clinical Immunology Unit, Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Laura Buysrogge
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Patrick Matthys
- Laboratory of Immunobiology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Philippe E. Van den Steen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Cui Y, Hackett RG, Ascue J, Muralidaran V, Patil D, Kang J, Kaufman SS, Khan K, Kroemer A. Innate and Adaptive Immune Responses in Intestinal Transplant Rejection: Through the Lens of Inflammatory Bowel and Intestinal Graft-Versus-Host Diseases. Gastroenterol Clin North Am 2024; 53:359-382. [PMID: 39068000 DOI: 10.1016/j.gtc.2024.01.002] [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: 07/30/2024]
Abstract
Intestinal transplantation is a life-saving procedure utilized for patients failing total parenteral nutrition. However, intestinal transplantattion remains plagued with low survival rates and high risk of allograft rejection. The authors explore roles of innate (macrophages, natural killer cells, innate lymphoid cells) and adaptive immune cells (Th1, Th2, Th17, Tregs) in inflammatory responses, particularly inflammatory bowel disease and graft versus host disease, and correlate these findings to intestinal allograft rejection, highlighting which effectors exacerbate or suppress intestinal rejection. Better understanding of this immunology can open further investigation into potential biomolecular targets to develop improved therapeutic treatment options and immunomonitoring techniques to combat allograft rejection and enhance patient lives.
Collapse
Affiliation(s)
- Yuki Cui
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Ryan G Hackett
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Jhalen Ascue
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Vinona Muralidaran
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Digvijay Patil
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Jiman Kang
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA; Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, USA
| | - Stuart S Kaufman
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Khalid Khan
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Alexander Kroemer
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
6
|
Swaminathan S, Mai LT, Meli AP, Carmona-Pérez L, Charpentier T, Lamarre A, King IL, Stäger S. LAG-3- and CXCR5-expressing CD4 T cells display progenitor-like properties during chronic visceral leishmaniasis. Cell Rep 2024; 43:113879. [PMID: 38416647 DOI: 10.1016/j.celrep.2024.113879] [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: 08/16/2023] [Revised: 01/04/2024] [Accepted: 02/13/2024] [Indexed: 03/01/2024] Open
Abstract
Maintenance of CD4 T cells during chronic infections is vital for limiting pathogen burden and disease recrudescence. Although inhibitory receptor expression by CD4 T cells is commonly associated with immune suppression and exhaustion, such cell-intrinsic mechanisms that control activation are also associated with cell survival. Using a mouse model of visceral leishmaniasis (VL), we discovered a subset of lymphocyte activation gene 3 (LAG-3)-expressing CD4 T cells that co-express CXCR5. Although LAG3+CXCR5+ CD4 T cells are present in naive mice, they expand during VL. These cells express gene signatures associated with self-renewal capacity, suggesting progenitor-like properties. When transferred into Rag1-/- mice, these LAG3+CXCR5+ CD4 T cells differentiated into multiple effector types upon Leishmania donovani infection. The transcriptional repressor B cell lymphoma-6 was partially required for their maintenance. Altogether, we propose that the LAG3+CXCR5+ CD4 T cell subset could play a role in maintaining CD4 T cell responses during persistent infections.
Collapse
Affiliation(s)
- Sharada Swaminathan
- INRS-Centre Armand-Frappier Santé Biotechnologie and Infectiopôle INRS, 531 Boulevard des Prairies, Laval, QC, Canada
| | - Linh Thuy Mai
- INRS-Centre Armand-Frappier Santé Biotechnologie and Infectiopôle INRS, 531 Boulevard des Prairies, Laval, QC, Canada
| | - Alexandre P Meli
- Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, Meakins-Christie Laboratories, McGill Centre for Microbiome Research, McGill University, Montreal, QC, Canada
| | - Liseth Carmona-Pérez
- INRS-Centre Armand-Frappier Santé Biotechnologie and Infectiopôle INRS, 531 Boulevard des Prairies, Laval, QC, Canada
| | - Tania Charpentier
- INRS-Centre Armand-Frappier Santé Biotechnologie and Infectiopôle INRS, 531 Boulevard des Prairies, Laval, QC, Canada
| | - Alain Lamarre
- INRS-Centre Armand-Frappier Santé Biotechnologie and Infectiopôle INRS, 531 Boulevard des Prairies, Laval, QC, Canada
| | - Irah L King
- Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, Meakins-Christie Laboratories, McGill Centre for Microbiome Research, McGill University, Montreal, QC, Canada
| | - Simona Stäger
- INRS-Centre Armand-Frappier Santé Biotechnologie and Infectiopôle INRS, 531 Boulevard des Prairies, Laval, QC, Canada.
| |
Collapse
|
7
|
Chen Q, Wei T, Li M, Liu S, Wu J, Xu G, Zou J, Xie S. Effect of aqueous extract of Millettia speciosa Champ on intestinal health maintenance and immune enhancement of Cyprinus carpio. FISH & SHELLFISH IMMUNOLOGY 2024; 144:109227. [PMID: 37984616 DOI: 10.1016/j.fsi.2023.109227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Abstract
Millettia speciosa Champ (MSP) is a natural Chinese herb that improves gastrointestinal health and enhances animal immunity. An 8-week feeding trial with different MSP levels (0, 150, 300, and 600 mg/kg) was conducted to evaluate the promotive effects of MSP in Cyprinus carpio. Results indicate that MSP improved intestinal immunity to some extent evidenced by the immuno-antioxidant parameters and the 16S rRNA in the Illumina MiSeq platform. With the analysis of transcriptome sequencing, 4685 differentially expressed genes (DEGs) were identified, including 2149 up-regulated and 2536 down-regulated. According to the GO and KEGG enrichments, DEGs were mainly involved in the immune system. Transcriptional expression of the NOD-like signaling pathway and key genes retrieved from the transcriptome database confirmed that innate immunity was improved in response to dietary MSP administration. Therefore, MSP could be used as a feed supplement that enhances immunity. This may provide insight into Chinese herb additive application in aquaculture production.
Collapse
Affiliation(s)
- Qingshi Chen
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Tianli Wei
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Min Li
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Shulin Liu
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Jinxia Wu
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance, Xiamen University, Xiamen, Fujian, 361005, China
| | - Guohuan Xu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Jixing Zou
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China.
| | - Shaolin Xie
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
8
|
Jiang H, Jiang J. Balancing act: the complex role of NK cells in immune regulation. Front Immunol 2023; 14:1275028. [PMID: 38022497 PMCID: PMC10652757 DOI: 10.3389/fimmu.2023.1275028] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Natural killer (NK) cells, as fundamental components of innate immunity, can quickly react to abnormalities within the body. In-depth research has revealed that NK cells possess regulatory functions not only in innate immunity but also in adaptive immunity under various conditions. Multiple aspects of the adaptive immune process are regulated through NK cells. In our review, we have integrated multiple studies to illuminate the regulatory function of NK cells in regulating B cell and T cell responses during adaptive immune processes, focusing on aspects including viral infections and the tumor microenvironment (TME). These insights provide us with many new understandings on how NK cells regulate different phases of the adaptive immune response.
Collapse
Affiliation(s)
- Hongwei Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute for Cell Therapy, Soochow University, Changzhou, Jiangsu, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute for Cell Therapy, Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
9
|
Rim S, Sakkestad ST, Zhou F, Gullaksen SE, Skavland J, Chauhan SK, Steinsland H, Hanevik K. Dynamics of circulating lymphocytes responding to human experimental enterotoxigenic Escherichia coli infection. Eur J Immunol 2023; 53:e2250254. [PMID: 37102399 DOI: 10.1002/eji.202250254] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/11/2023] [Accepted: 04/24/2023] [Indexed: 04/28/2023]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is an important cause of children's and travelers' diarrhea, with no licensed vaccine. This study aimed to explore the role of cellular immunity in protection against human ETEC infection. Nine volunteers were experimentally infected with ETEC, of which six developed diarrhea. Lymphocytes were collected from peripheral blood buffy coats, before and 3, 5, 6, 7, 10, and 28 days after dose ingestion, and 34 phenotypic and functional markers were examined by mass cytometry. Thirty-three cell populations, derived by manually merging 139 cell clusters from the X-shift unsupervised clustering algorithm, were analyzed. Initially, the diarrhea group responded with increased CD56dim CD16+ natural killer cells, dendritic cells tended to rise, and mucosal-associated invariant T cells decreased. On day 5-7, an increase in plasmablasts was paralleled by a consistent rise in CD4+ Th17-like effector memory and regulatory cell subsets. CD4+ Th17-like central memory cells peaked on day 10. All Th17-like cell populations showed increased expression of activation, gut-homing, and proliferation markers. Interestingly, in the nondiarrhea group, these same CD4+ Th17-like cell populations expanded earlier, normalizing around day 7. Earlier development of these CD4+ Th17-like cell populations in the nondiarrhea group may suggest a recall response and a potential role in controlling ETEC infections.
Collapse
Affiliation(s)
- Sehee Rim
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Sunniva T Sakkestad
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Fan Zhou
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Stein-Erik Gullaksen
- Department of Clinical Science, Centre of Cancer Biomarkers (CCBIO), University of Bergen, Bergen, Norway
- Hematology Section, Department of Internal Medicine, Helse Bergen, Bergen, Norway
| | - Jørn Skavland
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Sudhir K Chauhan
- Division of Cancer Medicine, Department of Cancer Immunology, Oslo University Hospital, Oslo, Norway
| | - Hans Steinsland
- Department of Global Public Health and Primary Care, Faculty of Medicine, Centre for Intervention Science in Maternal and Child Health (CISMAC), Centre for International Health, University of Bergen, Bergen, Norway
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Kurt Hanevik
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Medicine, Norwegian National Advisory Unit on Tropical Infectious Diseases, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
10
|
Mansourabadi AH, Aghamajidi A, Dorfaki M, Keshavarz F, Shafeghat Z, Moazzeni A, Arab FL, Rajabian A, Roozbehani M, Falak R, Faraji F, Jafari R. B lymphocytes in COVID-19: a tale of harmony and discordance. Arch Virol 2023; 168:148. [PMID: 37119286 PMCID: PMC10147999 DOI: 10.1007/s00705-023-05773-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/29/2023] [Indexed: 05/01/2023]
Abstract
B lymphocytes play a vital role in the human defense against viral infections by producing specific antibodies. They are also critical for the prevention of infectious diseases by vaccination, and their activation influences the efficacy of the vaccination. Since the beginning of coronavirus disease 2019 (COVID-19), which became the main concern of the world health system, many efforts have been made to treat and prevent the disease. However, for the development of successful therapeutics and vaccines, it is necessary to understand the interplay between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, and the immune system. The innate immune system provides primary and nonspecific defense against the virus, but within several days after infection, a virus-specific immune response is provided first by antibody-producing B cells, which are converted after the resolution of disease to memory B cells, which provide long-term immunity. Although a failure in B cell activation or B cell dysfunction can cause a severe form of the disease and also lead to vaccination inefficiency, some individuals with B cell immunodeficiency have shown less production of the cytokine IL-6, resulting in a better disease outcome. In this review, we present the latest findings on the interaction between SARS-CoV-2 and B lymphocytes during COVID-19 infection.
Collapse
Affiliation(s)
- Amir Hossein Mansourabadi
- Department of Immunology, School of medicine, Tehran University of Medical Sciences, Tehran, Iran
- Immunogenetics Research Network (IgReN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Azin Aghamajidi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Dorfaki
- Department of Microbiology and Immunology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Keshavarz
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Shafeghat
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Moazzeni
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Fahimeh Lavi Arab
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Research Center, School of Medicine, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arezoo Rajabian
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Roozbehani
- Vaccine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Faraji
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
- Institue of Immunology and Infectious diseases, Hazrat-e Rasool General Hospital, Floor 3, Building no. 3, Niyayesh St, Sattar Khan St, 1445613131, Tehran, Iran.
| | - Reza Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.
- Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Shafa St., Ershad Blvd, P.O. Box: 1138, 57147, Urmia, Iran.
| |
Collapse
|
11
|
Ott LC, Cuenca AG. Innate immune cellular therapeutics in transplantation. FRONTIERS IN TRANSPLANTATION 2023; 2:1067512. [PMID: 37994308 PMCID: PMC10664839 DOI: 10.3389/frtra.2023.1067512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Successful organ transplantation provides an opportunity to extend the lives of patients with end-stage organ failure. Selectively suppressing the donor-specific alloimmune response, however, remains challenging without the continuous use of non-specific immunosuppressive medications, which have multiple adverse effects including elevated risks of infection, chronic kidney injury, cardiovascular disease, and cancer. Efforts to promote allograft tolerance have focused on manipulating the adaptive immune response, but long-term allograft survival rates remain disappointing. In recent years, the innate immune system has become an attractive therapeutic target for the prevention and treatment of transplant organ rejection. Indeed, contemporary studies demonstrate that innate immune cells participate in both the initial alloimmune response and chronic allograft rejection and undergo non-permanent functional reprogramming in a phenomenon termed "trained immunity." Several types of innate immune cells are currently under investigation as potential therapeutics in transplantation, including myeloid-derived suppressor cells, dendritic cells, regulatory macrophages, natural killer cells, and innate lymphoid cells. In this review, we discuss the features and functions of these cell types, with a focus on their role in the alloimmune response. We examine their potential application as therapeutics to prevent or treat allograft rejection, as well as challenges in their clinical translation and future directions for investigation.
Collapse
Affiliation(s)
- Leah C Ott
- Department of General Surgery, Boston Children's Hospital, Boston, MA, United States
| | - Alex G Cuenca
- Department of General Surgery, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
12
|
Riddell P, Juvet SC. Natural killer cells, CMV infection, and antibody-mediated rejection. J Heart Lung Transplant 2023; 42:315-316. [PMID: 36804059 DOI: 10.1016/j.healun.2022.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Affiliation(s)
- P Riddell
- Irish Lung Transplant Program, Mater Misericordiae University Hospital, Dublin, Ireland.
| | - S C Juvet
- Toronto Lung Transplant Program, Toronto General Hospital, Toronto, Canada
| |
Collapse
|
13
|
Deng Y, Shi S, Luo J, Zhang Y, Dong H, Wang X, Zhou J, Wei Z, Li J, Xu C, Xu S, Sun Y, Ni B, Wu Y, Yang D, Han C, Tian Y. Regulation of mRNA stability contributes to the function of innate lymphoid cells in various diseases. Front Immunol 2023; 14:1118483. [PMID: 36776864 PMCID: PMC9909350 DOI: 10.3389/fimmu.2023.1118483] [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: 12/07/2022] [Accepted: 01/16/2023] [Indexed: 01/28/2023] Open
Abstract
Innate lymphoid cells (ILCs) are important subsets of innate immune cells that regulate mucosal immunity. ILCs include natural killer cells, innate lymphoid cells-1 (ILC1s), ILC2s, and ILC3s, which have extremely important roles in the immune system. In this review, we summarize the regulation of mRNA stability mediated through various factors in ILCs (e.g., cytokines, RNA-binding proteins, non-coding RNAs) and their roles in mediating functions in different ILC subsets. In addition, we discuss potential therapeutic targets for diseases such as chronic obstructive pulmonary disease, cancer, and pulmonary fibrosis by regulation of mRNA stability in ILCs, which may provide novel directions for future clinical research.
Collapse
Affiliation(s)
- Yuanyu Deng
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Saiyu Shi
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jie Luo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yiwei Zhang
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hui Dong
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xian Wang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong, China
| | - Jian Zhou
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhiyuan Wei
- The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiahui Li
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chen Xu
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shuai Xu
- Department of Stomatology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yi Sun
- The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, China
| | - Bing Ni
- Department of Pathophysiology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuzhang Wu
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Di Yang
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China,*Correspondence: Yi Tian, ; Di Yang, ; Chao Han,
| | - Chao Han
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China,*Correspondence: Yi Tian, ; Di Yang, ; Chao Han,
| | - Yi Tian
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China,*Correspondence: Yi Tian, ; Di Yang, ; Chao Han,
| |
Collapse
|
14
|
Liang C, Li S, Yuan J, Song Y, Ren W, Wang W, Shang Y, Tang S, Pang Y. Attenuated Cytokine-Induced Memory-Like Natural Killer Cell Responses to Mycobacterium tuberculosis in Tuberculosis Patients. Infect Drug Resist 2023; 16:2349-2364. [PMID: 37101462 PMCID: PMC10124624 DOI: 10.2147/idr.s407742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/15/2023] [Indexed: 04/28/2023] Open
Abstract
Purpose This study aimed to investigate the phenotype, proliferation and functional alterations of cytokine-induced memory-like natural killer (CIML NK) cells from healthy subjects and TB patients, and assessed the efficacy of CIML NK cells in response to H37Rv-infected U937 cells in vitro. Methods Fresh peripheral blood mononuclear cells (PBMCs) were isolated from healthy people and tuberculosis patients and activated for 16h using low-dose IL-15, or IL-12, IL-15, IL-18 combination or IL-12, IL-15, IL-18 and MTB H37Rv lysates, respectively, followed by low-dose IL-15 maintenance for another 7 days. Then, the PBMCs were co-cultured with K562 and H37Rv-infected U937, and the purified NK cells were co-cultured with H37Rv infected U937. The phenotype, proliferation and response function of CIML NK cells were assessed using flow cytometry. Finally, colony forming units were enumerated to confirm the survival of intracellular MTB. Results CIML NK phenotypes from TB patients were similar to healthy controls. CIML NK cells undergo higher rates of proliferation after IL-12/15/18 pre-activation. Moreover, the poor expansion potential of CIML NK cells co-stimulated with MTB lysates. CIML NK cells from healthy individuals showed enhanced IFN-γ functional to H37Rv infected U937 cells, along with significantly enhanced killing of H37Rv. However, the CIML NK cells from TB patients show attenuated IFN-γ production and now enhanced the ability of killing intracellular MTB compared to those from healthy donors after co-cultured with H37Rv infected U937. Conclusion CIML NK cells from healthy individuals exist the increased ability of IFN-γ secretion and boosted anti-MTB activity in vitro, which from TB patients show impaired IFN-γ production and no enhanced anti-MTB activity compared to those from healthy donors. Additionally, we observe the poor expansion potential of CIML NK cells co-stimulated with antigens from MTB. These results open up new possibilities for NK cell-based anti-tuberculosis immunotherapeutic strategies.
Collapse
Affiliation(s)
- Chen Liang
- Department of Bacteriology and Immunology, Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University /Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, People’s Republic of China
- Tuberculosis Clinical Medical Center, Beijing Chest Hospital, Capital Medical University /Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, People’s Republic of China
| | - Shanshan Li
- Department of Bacteriology and Immunology, Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University /Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, People’s Republic of China
| | - Jinfeng Yuan
- Department of Bacteriology and Immunology, Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University /Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, People’s Republic of China
| | - Yanhua Song
- Department of Tuberculosis, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, People’s Republic of China
| | - Weicong Ren
- Department of Bacteriology and Immunology, Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University /Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, People’s Republic of China
| | - Wei Wang
- Department of Bacteriology and Immunology, Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University /Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, People’s Republic of China
| | - Yuanyuan Shang
- Department of Bacteriology and Immunology, Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University /Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, People’s Republic of China
| | - Shenjie Tang
- Tuberculosis Clinical Medical Center, Beijing Chest Hospital, Capital Medical University /Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, People’s Republic of China
- Correspondence: Shenjie Tang; Yu Pang, Beijing Chest Hospital, Capital Medical University, No. 97, Machang, Tongzhou District, Beijing, 101149, People’s Republic of China, Tel/Fax +86 010 8950 9367; +86 010 8950 9359, Email ;
| | - Yu Pang
- Department of Bacteriology and Immunology, Beijing Key Laboratory on Drug-Resistant Tuberculosis Research, Beijing Chest Hospital, Capital Medical University /Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, People’s Republic of China
| |
Collapse
|
15
|
Hosseinzadeh F, Ai J, Hajifathali A, Muhammadnejad S, Ebrahimi-Barough S, Seyhoun I, Komeili Movahed T, Shirian S, Hosseinzadeh F, Ahmadpour S, Alijani M, Verdi J. The effects of Sorafenib and Natural killer cell co-injection in combinational treatment of hepatocellular carcinoma; an in vivo approach. Pharmacol Rep 2022; 74:379-391. [PMID: 35089543 DOI: 10.1007/s43440-021-00335-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 10/16/2021] [Accepted: 10/19/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Natural killer cells (NKC) and Sorafenib (Sor) are two important agents for the treatment of hepatocellular carcinoma (HCC). Over the past decade, the interaction of Sor and NKC against HCC has been widely challenging. This study aimed to assess the efficacy of NKC & Sor for the treatment of HCC in vivo. METHODS Subcutaneous xenograft models of HCC were established in nude mice. For safety assessment of treatment, the kidney and liver functions were analyzed. Paraffin embedded tumor sections were histopathologically studied and immunohistochemistry (IHC) tests were done to evaluate the angiogenesis (CD34) and proliferation (Ki67) indexes. The terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay was performed to identify the tumor cells undergoing apoptosis. The serum levels of tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) were measured by enzyme-linked immunosorbent assay (ELISA) and expression levels of major inflammatory cytokines and cytoplasmic granules in xenograft HCC were quantified using real-time PCR. RESULTS NKC & Sor significantly inhibited necrosis and apoptosis in tumor cells and increased angiogenesis and proliferation of HCC compared to the monotherapy of NKC or Sor alone. The serum levels of TNF-α, IFN-γ as well as the expression levels of TNF-α, IFN-γ, interleukins (ILs)-1, 6, 10, granzyme-B and perforin in the xenograft HCC tissues of the treated mice with NKC & Sor were significantly lower than those of treated with NKC or Sor alone. CONCLUSION Therapy with the specific dosage of NKC & Sor could not inhibit the HCC xenograft growth rate through a synergistic effect in a mouse model of HCC.
Collapse
Affiliation(s)
- Faezeh Hosseinzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran. .,Department of Tissue Engineering, Qom University of Medical Sciences, Qom, Iran. .,Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
| | - Jafar Ai
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samad Muhammadnejad
- Gene Therapy Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Iman Seyhoun
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sadegh Shirian
- Department of Pathology, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran.,Shiraz Molecular Pathology Research Center, Dr. Daneshbod Path Lab, Shiraz, Iran
| | | | - Sajjad Ahmadpour
- Gastroenterology and Hepatology Diseases Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Mohammadreza Alijani
- Department of Pathology, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran.,Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Javad Verdi
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Lian G, Mak TSK, Yu X, Lan HY. Challenges and Recent Advances in NK Cell-Targeted Immunotherapies in Solid Tumors. Int J Mol Sci 2021; 23:164. [PMID: 35008589 PMCID: PMC8745474 DOI: 10.3390/ijms23010164] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/08/2021] [Accepted: 12/20/2021] [Indexed: 02/06/2023] Open
Abstract
Natural killer (NK) cell is a powerful malignant cells killer, providing rapid immune responses via direct cytotoxicity without the need of antigen processing and presentation. It plays an essential role in preventing early tumor, metastasis and minimal residual disease. Although adoptive NK therapies achieved great success in clinical trials against hematologic malignancies, their accumulation, activation, cytotoxic and immunoregulatory functions are severely impaired in the immunosuppressive microenvironment of solid tumors. Now with better understandings of the tumor evasive mechanisms from NK-mediated immunosurveillance, immunotherapies targeting the key molecules for NK cell dysfunction and exhaustion have been developed and tested in both preclinical and clinical studies. In this review, we introduce the challenges that NK cells encountered in solid tumor microenvironment (TME) and the therapeutic approaches to overcome these limitations, followed by an outline of the recent preclinical advances and the latest clinical outcomes of NK-based immunotherapies, as well as promising strategies to optimize current NK-targeted immunotherapies for solid tumors.
Collapse
Affiliation(s)
- Guangyu Lian
- Guangdong-Hong Kong Joint Research Laboratory on Immunological and Genetic Kidney Diseases, Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China;
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Department of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China;
| | - Thomas Shiu-Kwong Mak
- Department of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China;
| | - Xueqing Yu
- Guangdong-Hong Kong Joint Research Laboratory on Immunological and Genetic Kidney Diseases, Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China;
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Hui-Yao Lan
- Department of Medicine & Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China;
| |
Collapse
|
17
|
Martinez-Espinosa I, Serrato JA, Ortiz-Quintero B. Role of IL-10-Producing Natural Killer Cells in the Regulatory Mechanisms of Inflammation during Systemic Infection. Biomolecules 2021; 12:biom12010004. [PMID: 35053151 PMCID: PMC8773486 DOI: 10.3390/biom12010004] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/08/2021] [Accepted: 12/11/2021] [Indexed: 12/29/2022] Open
Abstract
Natural killer (NK) cells have the dual ability to produce pro-inflammatory (IFNγ) and anti-inflammatory (IL-10) cytokines during systemic infection, which points to their crucial role both as inflammatory effectors for infection clearance and as regulators to counterbalance inflammation to limit immune-mediated damage to the host. In particular, immunosuppressive IL-10 secretion by NK cells has been described to occur in systemic, but not local, infections as a recent immunoregulatory mechanism of inflammation that may be detrimental or beneficial, depending on the timing of release, type of disease, or the infection model. Understanding the factors that drive the production of IL-10 by NK cells and their impact during dualistic inflammatory states, such as sepsis and other non-controlled inflammatory diseases, is relevant for achieving effective therapeutic advancements. In this review, the evidence regarding the immunoregulatory role of IL-10-producing NK cells in systemic infection is summarized and discussed in detail, and the potential molecular mechanisms that drive IL-10 production by NK cells are considered.
Collapse
|
18
|
Romano A, Brown N, Ashwin H, Doehl JSP, Hamp J, Osman M, Dey N, Rani GF, Ferreira TR, Kaye PM. Interferon-γ-Producing CD4 + T Cells Drive Monocyte Activation in the Bone Marrow During Experimental Leishmania donovani Infection. Front Immunol 2021; 12:700501. [PMID: 34557190 PMCID: PMC8453021 DOI: 10.3389/fimmu.2021.700501] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/18/2021] [Indexed: 12/24/2022] Open
Abstract
Ly6Chi inflammatory monocytes develop in the bone marrow and migrate to the site of infection during inflammation. Upon recruitment, Ly6Chi monocytes can differentiate into dendritic cells or macrophages. According to the tissue environment they can also acquire different functions. Several studies have described pre-activation of Ly6Chi monocytes in the bone marrow during parasitic infection, but whether this process occurs during experimental visceral leishmaniasis and, if so, the mechanisms contributing to their activation are yet to be established. In wild type C57BL/6 (B6) mice infected with Leishmania donovani, the number of bone marrow Ly6Chi monocytes increased over time. Ly6Chi monocytes displayed a highly activated phenotype from 28 days to 5 months post infection (p.i), with >90% expressing MHCII and >20% expressing iNOS. In comparison, in B6.Rag2-/- mice <10% of bone marrow monocytes were MHCII+ at day 28 p.i., an activation deficiency that was reversed by adoptive transfer of CD4+ T cells. Depletion of CD4+ T cells in B6 mice and the use of mixed bone marrow chimeras further indicated that monocyte activation was driven by IFNγ produced by CD4+ T cells. In B6.Il10-/- mice, L. donovani infection induced a faster but transient activation of bone marrow monocytes, which correlated with the magnitude of CD4+ T cell production of IFNγ and resolution of the infection. Under all of the above conditions, monocyte activation was associated with greater control of parasite load in the bone marrow. Through reinfection studies in B6.Il10-/- mice and drug (AmBisome®) treatment of B6 mice, we also show the dependence of monocyte activation on parasite load. In summary, these data demonstrate that during L. donovani infection, Ly6Chi monocytes are primed in the bone marrow in a process driven by CD4+ T cells and whereby IFNγ promotes and IL-10 limits monocyte activation and that the presence of parasites/parasite antigen plays a crucial role in maintaining bone marrow monocyte activation.
Collapse
Affiliation(s)
- Audrey Romano
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Najmeeyah Brown
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Helen Ashwin
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Johannes S P Doehl
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Jonathan Hamp
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Mohamed Osman
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Nidhi Dey
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Gulab Fatima Rani
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Tiago Rodrigues Ferreira
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Paul M Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| |
Collapse
|
19
|
Shmeleva EV, Colucci F. Maternal natural killer cells at the intersection between reproduction and mucosal immunity. Mucosal Immunol 2021; 14:991-1005. [PMID: 33903735 PMCID: PMC8071844 DOI: 10.1038/s41385-020-00374-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/24/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023]
Abstract
Many maternal immune cells populate the decidua, which is the mucosal lining of the uterus transformed during pregnancy. Here, abundant natural killer (NK) cells and macrophages help the uterine vasculature adapt to fetal demands for gas and nutrients, thereby supporting fetal growth. Fetal trophoblast cells budding off the forming placenta and invading deep into maternal tissues come into contact with these and other immune cells. Besides their homeostatic functions, decidual NK cells can respond to pathogens during infection, but in doing so, they may become conflicted between destroying the invader and sustaining fetoplacental growth. We review how maternal NK cells balance their double duty both in the local microenvironment of the uterus and systemically, during toxoplasmosis, influenza, cytomegalovirus, malaria and other infections that threat pregnancy. We also discuss recent developments in the understanding of NK-cell responses to SARS-Cov-2 infection and the possible dangers of COVID-19 during pregnancy.
Collapse
Affiliation(s)
- Evgeniya V Shmeleva
- Department of Obstetrics & Gynaecology, University of Cambridge, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, CB2 0SW, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Francesco Colucci
- Department of Obstetrics & Gynaecology, University of Cambridge, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, CB2 0SW, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
20
|
New insights into regulatory B cells biology in viral, bacterial, and parasitic infections. INFECTION GENETICS AND EVOLUTION 2021; 89:104753. [PMID: 33545392 DOI: 10.1016/j.meegid.2021.104753] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/16/2021] [Accepted: 01/29/2021] [Indexed: 12/14/2022]
Abstract
B lymphocytes are primarily well known for their contribution to immunity by antibody production, antigen presentation and, the production of cytokines. In recent years several studies demonstrated the existence of B cells with regulatory functions, which have been termed regulatory B cells (Bregs), similar to regulatory T cells (Tregs). Bregs are a subpopulation of B cells that have immunosuppressive effects via the production of regulatory cytokines including interleukin-10 (IL-10), transforming growth factor-β (TGF-β), and IL-35. Bregs limit host defense against various pathogens. In addition, Bregs contribute to increased levels of regulatory cytokines and leads to an induction of suppressive Tregs, which exert broader suppressive functions against various pathogens. The high percentage of Bregs is positively associated with viral and bacterial load and can contribute to poor vaccine responses. Bregs can also facilitate pathogen survival at an early stage of infection, and subsequently cause increased severity of disease by inhibiting pro-inflammatory cytokine production, macrophage activation, and inflammatory T cells activation such as Th1, Th17, and Th22. Also, Bregs afford protection against the hyper-inflammatory response in parasitic infections. Here we review the central role of Bregs in many major bacterial and viral human infections, and provide an overview of the immunoregulatory mechanisms used by Bregs.
Collapse
|
21
|
Clark SE, Schmidt RL, Aguilera ER, Lenz LL. IL-10-producing NK cells exacerbate sublethal Streptococcus pneumoniae infection in the lung. Transl Res 2020; 226:70-82. [PMID: 32634590 PMCID: PMC7572800 DOI: 10.1016/j.trsl.2020.07.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/27/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023]
Abstract
Lung inflammation is tightly controlled to balance microbial clearance with the tissue damage that accompanies this response. Bacterial pathogens including Streptococcus pneumoniae (S. pneumoniae) modulate immune regulation by promoting secretion of the anti-inflammatory cytokine IL-10. The important cellular sources of IL-10 that impact protection against different bacterial infections are not well characterized. We find that S. pneumoniaeactivates IL-10 secretion from natural killer (NK) cells in the lung, which restrict host protection in a mouse model of sublethal infection. Direct transfer of wild-type NK cells into the lungs of IL-10-deficient mice drives bacterial expansion, identifying NK cells as a critical source of IL-10 promoting S. pneumoniae infection. The S. pneumoniae virulence protein Spr1875 was found to elicit NK cell IL-10 production in purified cells and in the lungs of live animals. These findings reveal therapeutic targets to combat bacterial-driven immune regulation in the lung.
Collapse
Affiliation(s)
- Sarah E Clark
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.
| | - Rebecca L Schmidt
- Department of Biomedical Sciences, National Jewish Health, Denver, Colorado; Department of Biology and Chemistry, Upper Iowa University, Fayette, Iowa
| | - Elizabeth R Aguilera
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado
| | - Laurel L Lenz
- Department of Biomedical Sciences, National Jewish Health, Denver, Colorado; Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
22
|
Zwirner NW, Domaica CI, Fuertes MB. Regulatory functions of NK cells during infections and cancer. J Leukoc Biol 2020; 109:185-194. [PMID: 33095941 DOI: 10.1002/jlb.3mr0820-685r] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 08/16/2020] [Accepted: 09/24/2020] [Indexed: 01/02/2023] Open
Abstract
After recognition, NK cells can kill susceptible target cells through perforin-dependent mechanisms or by inducing death receptor-mediated apoptosis, and they can also secrete cytokines that are pivotal for immunomodulation. Despite the critical role as effector cells against tumors and virus-infected cells, NK cells have been implicated in the regulation of T cell-mediated responses in different models of autoimmunity, transplantation, and viral infections. Here, we review the mechanisms described for NK cell-mediated inhibition of adaptive immune responses, with spotlight on the emerging evidence of their regulatory role that shapes antitumor immune responses.
Collapse
Affiliation(s)
- Norberto W Zwirner
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carolina I Domaica
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Mercedes B Fuertes
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
23
|
van der Heide SL, Xi Y, Upham JW. Natural Killer Cells and Host Defense Against Human Rhinoviruses Is Partially Dependent on Type I IFN Signaling. Front Cell Infect Microbiol 2020; 10:510619. [PMID: 33194777 PMCID: PMC7609819 DOI: 10.3389/fcimb.2020.510619] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 10/02/2020] [Indexed: 12/12/2022] Open
Abstract
Rhinovirus (RV), the causative agent of the common cold, causes only mild upper respiratory tract infections in healthy individuals, but can cause longer lasting and more severe pulmonary infections in people with chronic lung diseases and in the setting of immune suppression or immune deficiency. RV-infected lung structural cells release type I interferon (IFN-I), initiating the immune response, leading to protection against viruses in conjunction with migratory immune cells. However, IFN-I release is deficient in some people with asthma. Innate immune cells, such as natural killer (NK) cells, are proposed to play major roles in the control of viral infections, and may contribute to exacerbations of chronic lung diseases, such as asthma. In this study, we characterized the NK cell response to RV infection using an in vitro model of infection in healthy individuals, and determined the extent to which IFN-I signaling mediates this response. The results indicate that RV stimulation in vitro induces NK cell activation in healthy donors, leading to degranulation and the release of cytotoxic mediators and cytokines. IFN-I signaling was partly responsible for NK cell activation and functional responses to RV. Overall, our findings suggest the involvement of NK cells in the control of RV infection in healthy individuals. Further understanding of NK cell regulation may deepen our understanding of the mechanisms that contribute to susceptibility to RV infections in asthma and other chronic lung diseases.
Collapse
Affiliation(s)
- Saskia L van der Heide
- Lung and Allergy Research Centre, Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Yang Xi
- Lung and Allergy Research Centre, Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - John W Upham
- Lung and Allergy Research Centre, Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia.,Department of Respiratory Medicine, Princess Alexandra Hospital, Brisbane, QLD, Australia
| |
Collapse
|
24
|
CAR-NK Therapy: Translating a Novel Immunotherapy Treatment to Solid Organ Transplantation. Transplantation 2020; 104:1523-1524. [PMID: 32732821 DOI: 10.1097/tp.0000000000003267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
25
|
Zeng S, Xiao Z, Wang Q, Guo Y, He Y, Zhu Q, Zou Y. Strategies to achieve immune tolerance in allogeneic solid organ transplantation. Transpl Immunol 2020; 58:101250. [PMID: 31655110 DOI: 10.1016/j.trim.2019.101250] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/19/2019] [Accepted: 10/21/2019] [Indexed: 12/14/2022]
|
26
|
Mai LT, Smans M, Silva-Barrios S, Fabié A, Stäger S. IRF-5 Expression in Myeloid Cells Is Required for Splenomegaly in L. donovani Infected Mice. Front Immunol 2020; 10:3071. [PMID: 32038622 PMCID: PMC6985270 DOI: 10.3389/fimmu.2019.03071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/16/2019] [Indexed: 01/14/2023] Open
Abstract
Persistent Leishmania donovani infection is characterized by chronic inflammation, immune suppression, and splenomegaly. We have previously reported that the transcription factor interferon regulatory factor 5 (IRF-5) is largely responsible for inducing the inflammatory response and maintaining protective Th1 cells following L. donovani inoculation in mice. However, the cellular source responsible for these effects is yet unknown. In this study, we investigated the role of IRF-5 in myeloid cells during experimental visceral leishmaniasis (VL). First, we show that the LysM-Cre mouse model is not suited for investigating gene expression in splenic myeloid cells during experimental VL. Using the Cd11c-Cre mouse model, we demonstrate that Irf5 expression in CD11c+ cells (monocytes, dendritic cells, activated macrophages) is essential for inducing splenomegaly and for recruiting myeloid cells to the spleen, but it is not required for the development or maintenance of parasite-specific IFNγ-producing CD4 T cells. CD11c-specific Irf5 -/- mice are more resistant to L. donovani infection, suggesting that the induction of splenomegaly is detrimental to the host.
Collapse
Affiliation(s)
- Linh Thuy Mai
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Laval, QC, Canada
| | - Mélina Smans
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Laval, QC, Canada
| | - Sasha Silva-Barrios
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Laval, QC, Canada
| | - Aymeric Fabié
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Laval, QC, Canada
| | - Simona Stäger
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Laval, QC, Canada
| |
Collapse
|
27
|
De Trez C, Stijlemans B, Bockstal V, Cnops J, Korf H, Van Snick J, Caljon G, Muraille E, Humphreys IR, Boon L, Van Ginderachter JA, Magez S. A Critical Blimp-1-Dependent IL-10 Regulatory Pathway in T Cells Protects From a Lethal Pro-inflammatory Cytokine Storm During Acute Experimental Trypanosoma brucei Infection. Front Immunol 2020; 11:1085. [PMID: 32655552 PMCID: PMC7325990 DOI: 10.3389/fimmu.2020.01085] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/05/2020] [Indexed: 01/12/2023] Open
Abstract
In many infectious diseases, the immune response operates as a double-edged sword. While required for protective immunity, infection-induced inflammation can be detrimental if it is not properly controlled, causing collateral body damage and potentially leading to death. It is in this context that the potent anti-inflammatory cytokine interleukin-10 (IL-10) is required to dampen the pro-inflammatory immune response that hallmarks trypanosomosis. Effective control of this infection requires not just the action of antibodies specific for the parasite's variable surface glycoprotein (VSG) coat antigens, but also a pro-inflammatory immune response mediated mainly by IFNγ, TNF, and NO. However, strict control of inflammation is mandatory, as IL-10-deficient mice succumb from an unrestrained cytokine storm within 10 days of a Trypanosome brucei infection. The relevant cellular source of IL-10 and the associated molecular mechanisms implicated in its trypanosomosis associated production are poorly understood. Using an IL-10 reporter mouse strain (Vert-X), we demonstrate here that NK cells, CD8+ T cells and CD4+ T cells as well as B cells and plasma cells constitute potential cellular sources of IL-10 within the spleen and liver during acute infection. The IL-10 wave follows peak pro-inflammatory cytokine production, which accompanied the control of peak parasitemia. Similar results were observed following conventional experimental needle infection and physiological infections via T. brucei-infected tsetse flies. Our results show that conditional T cell-specific ablation of the IL-10 regulating Prdm1 gene (encoding for the Blimp-1 transcription factor), leads to an uncontrolled trypanosome-induced pro-inflammatory syndrome like the one observed in infected IL-10-deficient mice. This result indicates that the biological role of IL-10-derived from non-T cells, including NK cells, is of minor importance when considering host survival. The cytokine IL-27 that is also considered to be an IL-10 regulator, did not affect IL-10 production during infection. Together, these data suggest that T. brucei activates a Blimp-1-dependent IL-10 regulatory pathway in T cells that acts as a critical anti-inflammatory rheostat, mandatory for host survival during the acute phase of parasitemia.
Collapse
Affiliation(s)
- Carl De Trez
- Research Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Benoit Stijlemans
- Research Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium.,Myeloid Cell Immunology Laboratory, VIB Centre for Inflammation Research, Brussels, Belgium
| | - Viki Bockstal
- Research Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Jennifer Cnops
- Research Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Hannelie Korf
- Laboratory of Hepatology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Jacques Van Snick
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium.,Ludwig Cancer Research, Brussels Branch, Brussels, Belgium
| | - Guy Caljon
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Wilrijk, Belgium
| | - Eric Muraille
- Unité de Recherche en Biologie des Microorganismes, Laboratoire d'Immunologie et de Microbiologie, Université de Namur, Namur, Belgium.,Laboratoire de Parasitologie, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Ian R Humphreys
- Division of Infection and Immunity/Systems Immunity University Research Institute, Cardiff University, Cardiff, United Kingdom
| | | | - Jo A Van Ginderachter
- Research Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium.,Myeloid Cell Immunology Laboratory, VIB Centre for Inflammation Research, Brussels, Belgium
| | - Stefan Magez
- Research Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium.,Ghent University Global, Incheon, South Korea
| |
Collapse
|
28
|
Gyurova IE, Ali A, Waggoner SN. Natural Killer Cell Regulation of B Cell Responses in the Context of Viral Infection. Viral Immunol 2019; 33:334-341. [PMID: 31800366 DOI: 10.1089/vim.2019.0129] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Secretion of both neutralizing and nonneutralizing virus-specific antibodies by B cells is a key component of immune control of many virus infections and a critical benchmark of successful preventative vaccines. Natural killer (NK) cells also play a vital role in antiviral immune defense via cytolytic elimination of infected cells and production of proinflammatory antiviral cytokines. Accumulating evidence points to multifaceted crosstalk between NK cells and antiviral B cell responses that can determine virus elimination, pathogenesis of infection, and efficacy of vaccine-elicited protection. These outcomes are a result of both positive and negative influences of NK cells on the B cell responses, as well as canonical antiviral killing of infected B cells. On one hand, NK cell-derived cytokines such as interferon-gamma (IFN-γ) may promote B cell activation and enhance immunoglobulin production. In contrast, NK cell immunoregulatory killing of CD4 T cells can limit affinity maturation in germinal centers resulting in weak infection or vaccine induction of antiviral neutralizing antibodies. In this review, we will discuss these and other dueling contributions of NK cells to B cell responses during virus infection or vaccination.
Collapse
Affiliation(s)
- Ivayla E Gyurova
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Pathobiology and Molecular Medicine Graduate Program, University of Cincinnati, Cincinnati, Ohio, USA
| | - Ayad Ali
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Medical Scientist Training Program, University of Cincinnati, Cincinnati, Ohio, USA.,Graduate Program in Immunology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Stephen N Waggoner
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Pathobiology and Molecular Medicine Graduate Program, University of Cincinnati, Cincinnati, Ohio, USA.,Medical Scientist Training Program, University of Cincinnati, Cincinnati, Ohio, USA.,Graduate Program in Immunology, University of Cincinnati, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
29
|
Ali AK, Komal AK, Almutairi SM, Lee SH. Natural Killer Cell-Derived IL-10 Prevents Liver Damage During Sustained Murine Cytomegalovirus Infection. Front Immunol 2019; 10:2688. [PMID: 31803193 PMCID: PMC6873346 DOI: 10.3389/fimmu.2019.02688] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/31/2019] [Indexed: 11/25/2022] Open
Abstract
Natural Killer (NK) cells are lymphocytes of the innate immune response that play a vital role in controlling infections and cancer. Their pro-inflammatory role has been well-established; however, less is known about the regulatory functions of NK cells, in particular, their production of the anti-inflammatory cytokine IL-10. In this study, we investigated the immunoregulatory function of NK cells during MCMV infection and demonstrated that NK cells are major producers of IL-10 during the early stage of infection. To investigate the effect of NK cell-derived IL-10, we have generated NK cell-specific IL-10-deficient mice (NKp46-Cre-Il10fl/fl) displaying no signs of age-related spontaneous inflammation, with NK cells that show no detectable IL-10 production upon in vitro stimulation. In NKp46-Cre-Il10fl/fl mice, the levels of IL-10 and IFNγ, viral burdens and T cell activation were similar between NKp46-Cre-Il10fl/fl mice and their control littermates, suggesting that NK cell-derived IL-10 is dispensable during acute MCMV infection in immunocompetent hosts. In perforin-deficient mice that show a more sustained infection, NK cells produce more sustained levels of IL-10. By crossing NKp46-Cre-Il10fl/fl mice with perforin-deficient mice, we demonstrated that NK cell-derived IL-10 regulates T cell activation, prevents liver damage, and allows for better disease outcome. Taken together, NK cell-derived IL-10 can be critical in regulating the immune response during early phases of infection and therefore protecting the host from excessive immunopathology.
Collapse
Affiliation(s)
- Alaa Kassim Ali
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Amandeep Kaur Komal
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Saeedah Musaed Almutairi
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Botany and Microbiology Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Seung-Hwan Lee
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
30
|
Burrack KS, Hart GT, Hamilton SE. Contributions of natural killer cells to the immune response against Plasmodium. Malar J 2019; 18:321. [PMID: 31533835 PMCID: PMC6751859 DOI: 10.1186/s12936-019-2953-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 09/07/2019] [Indexed: 01/08/2023] Open
Abstract
Natural killer (NK) cells are important innate effector cells that are well described in their ability to kill virally-infected cells and tumors. However, there is increasing appreciation for the role of NK cells in the control of other pathogens, including intracellular parasites such as Plasmodium, the cause of malaria. NK cells may be beneficial during the early phase of Plasmodium infection—prior to the activation and expansion of antigen-specific T cells—through cooperation with myeloid cells to produce inflammatory cytokines like IFNγ. Recent work has defined how Plasmodium can activate NK cells to respond with natural cytotoxicity, and inhibit the growth of parasites via antibody-dependent cellular cytotoxicity mechanisms (ADCC). A specialized subset of adaptive NK cells that are negative for the Fc receptor γ chain have enhanced ADCC function and correlate with protection from malaria. Additionally, production of the regulatory cytokine IL-10 by NK cells prevents overt pathology and death during experimental cerebral malaria. Now that conditional NK cell mouse models have been developed, previous studies need to be reevaluated in the context of what is now known about other immune populations with similarity to NK cells (i.e., NKT cells and type I innate lymphoid cells). This brief review summarizes recent findings which support the potentially beneficial roles of NK cells during Plasmodium infection in mice and humans. Also highlighted are how the actions of NK cells can be explored using new experimental strategies, and the potential to harness NK cell function in vaccination regimens.
Collapse
Affiliation(s)
- Kristina S Burrack
- Department of Medicine, Hennepin Healthcare Research Institute, Minneapolis, MN, 55415, USA
| | - Geoffrey T Hart
- Center for Immunology, Department of Medicine, Division of Infectious Disease and International Medicine, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Sara E Hamilton
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55414, USA.
| |
Collapse
|
31
|
Clark SE, Burrack KS, Jameson SC, Hamilton SE, Lenz LL. NK Cell IL-10 Production Requires IL-15 and IL-10 Driven STAT3 Activation. Front Immunol 2019; 10:2087. [PMID: 31552035 PMCID: PMC6736993 DOI: 10.3389/fimmu.2019.02087] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 08/19/2019] [Indexed: 01/22/2023] Open
Abstract
Natural killer (NK) cells can produce IFNγ or IL-10 to regulate inflammation and immune responses but the factors driving NK cell IL-10 secretion are poorly-defined. Here, we identified NK cell-intrinsic STAT3 activation as vital for IL-10 production during both systemic Listeria monocytogenes (Lm) infection and following IL-15 cytokine/receptor complex (IL15C) treatment for experimental cerebral malaria (ECM). In both contexts, conditional Stat3 deficiency in NK cells abrogated production of IL-10. Initial NK cell STAT3 phosphorylation was driven by IL-15. During Lm infection, this required capture or presentation of IL-15 by NK cell IL-15Rα. Persistent STAT3 activation was required to drive measurable IL-10 secretion and required NK cell expression of IL-10Rα. Survival-promoting effects of IL-15C treatment in ECM were dependent on NK cell Stat3 while NK cell-intrinsic deficiency for Stat3, Il15ra, or Il10ra abrogated NK cell IL-10 production and increased resistance against Lm. NK cell Stat3 deficiency did not impact production of IFNγ, indicating the STAT3 activation initiated by IL-15 and amplified by IL-10 selectively drives the production of anti-inflammatory IL-10 by responding NK cells.
Collapse
Affiliation(s)
- Sarah E Clark
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Kristina S Burrack
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Stephen C Jameson
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Sara E Hamilton
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Laurel L Lenz
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
32
|
Beetz O, Kolb J, Buck B, Trautewig B, Timrott K, Vondran FWR, Meder I, Löbbert C, Hundrieser J, Klempnauer J, Bektaş H, Lieke T. Recipient natural killer cells alter the course of rejection of allogeneic heart grafts in rats. PLoS One 2019; 14:e0220546. [PMID: 31437165 PMCID: PMC6705777 DOI: 10.1371/journal.pone.0220546] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022] Open
Abstract
Rejection of solid organ grafts is regarded to be dependent on T cell responses. Nonetheless, numerous studies have focused on the contribution of NK cells in this process, resulting in contradictory theories. While some conclude that there is no participation of NK cells, others found an inflammatory or regulative role of NK cells. However, the experimental settings are rarely comparable with regard to challenged species, strain combinations or the nature of the graft. Thus, clear definition of NK cell contribution might be impeded by these circumstances. In this study we performed heterotopic heart transplantation (HTx) in rats, choosing one donor-recipient-combination leading to a fast and a second leading to a prolonged course of graft rejection. We intervened in the rejection process, by depletion of recipient NK cells on the one hand and by injection of activated NK cells syngeneic to the recipients on the other. The fast course of rejection could not be influenced by any of the NK cell manipulative treatments. However, the more prolonged course of rejection was highly susceptible to depletion of NK cells, resulting in significant acceleration of rejection, while injection of NK cells induced acceptance of the grafts. We suggest that, depending on the specific setting, NK cells can attenuate the first trigger of immune response, which allows establishing the regulatory activity leading to tolerance of the graft.
Collapse
Affiliation(s)
- Oliver Beetz
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Joline Kolb
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Benjamin Buck
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Britta Trautewig
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
- Transplant Laboratory, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Kai Timrott
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Florian W. R. Vondran
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Ingrid Meder
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Corinna Löbbert
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Joachim Hundrieser
- Transplant Laboratory, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Jürgen Klempnauer
- Transplant Laboratory, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Hüseyin Bektaş
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
- Department of General-, Visceral- and Oncological Surgery, Hospital Group Gesundheit Nord, Bremen, Germany
| | - Thorsten Lieke
- Regenerative Medicine and Experimental Surgery, Department of General-, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
33
|
McFarlane E, Mokgethi T, Kaye PM, Hurdayal R, Brombacher F, Alexander J, Carter KC. IL-4 Mediated Resistance of BALB/c Mice to Visceral Leishmaniasis Is Independent of IL-4Rα Signaling via T Cells. Front Immunol 2019; 10:1957. [PMID: 31475014 PMCID: PMC6707061 DOI: 10.3389/fimmu.2019.01957] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/02/2019] [Indexed: 01/10/2023] Open
Abstract
Previous studies infecting global IL-4Rα−/−, IL-4−/−, and IL-13−/−mice on a BALB/c background with the visceralizing parasite Leishmania donovani have shown that the T helper 2 cytokines, IL-4, and IL-13, play influential but not completely overlapping roles in controlling primary infection. Subsequently, using macrophage/neutrophil-specific IL-4Rα deficient BALB/c mice, we demonstrated that macrophage/neutrophil unresponsiveness to IL-4 and IL-13 did not have a detrimental effect during L. donovani infection. Here we expand on these findings and show that CD4+ T cell-(Lckcre), as well as pan T cell-(iLckcre) specific IL-4Rα deficient mice, on a BALB/c background, unlike global IL-4Rα deficient mice, are also not adversely affected in terms of resistance to primary infection with L. donovani. Our analysis suggested only a transient and tissue specific impact on disease course due to lack of IL-4Rα on T cells, limited to a reduced hepatic parasite burden at day 30 post-infection. Consequently, the protective role(s) demonstrated for IL-4 and IL-13 during L. donovani infection are mediated by IL-4Rα-responsive cell(s) other than macrophages, neutrophils and T cells.
Collapse
Affiliation(s)
- Emma McFarlane
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Thabang Mokgethi
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Paul M Kaye
- Department of Biology, Centre for Immunology and Infection, Hull York Medical School, University of York, York, United Kingdom
| | - Ramona Hurdayal
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine (IDM), South African Medical Research Council (SAMRC) on Immunology of Infectious Diseases, University of Cape Town, Cape Town, South Africa.,Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa.,Department of Molecular and Cell Biology, University of Cape Town, Cape Town, South Africa
| | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology, Cape Town Component, Cape Town, South Africa.,Division of Immunology, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine (IDM), South African Medical Research Council (SAMRC) on Immunology of Infectious Diseases, University of Cape Town, Cape Town, South Africa.,Faculty of Health Sciences, Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| | - James Alexander
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Katharine C Carter
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
34
|
Silva-Barrios S, Stäger S. Hypergammaglobulinemia sustains the development of regulatory responses during chronic Leishmania donovani infection in mice. Eur J Immunol 2019; 49:1082-1091. [PMID: 31001826 DOI: 10.1002/eji.201847917] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 03/01/2019] [Accepted: 04/04/2019] [Indexed: 01/19/2023]
Abstract
Visceral leishmaniasis, a chronic, potentially fatal disease, is characterized by high production of low-affinity antibodies. In humans, hypergammaglobulinemia is prediction of disease progression. Nevertheless, the contribution of hypermutated and/or class-switched immunoglobulins to disease pathogenesis has never been studied. Using Aicda-/- mice and the experimental model of Leishmania donovani infection, we demonstrate that the absence of hypermutated and/or class-switched antibodies was associated with increased resistance to disease, stronger protective Th1 responses, and a lower frequency of regulatory IFNγ+ IL-10+ CD4 T cells. Interestingly, stronger Th1 responses and the absence of IFNγ+ IL-10+ CD4 T cells during chronic infection in infected Aicda-/- mice were not caused by a T-cell intrinsic effect of AID, but by changes in the cytokine environment during chronic disease. Indeed TNF, IL-10 and IFN-ß expressions were only upregulated in the presence of hypermutated, class-switched antibodies and hypergammaglobulinemia at later stages of infection. Taken together, our results suggest that hypergammaglobulinemia sustains inhibitory responses during chronic visceral leishmaniasis.
Collapse
Affiliation(s)
| | - Simona Stäger
- INRS, Centre Armand-Frappier Santé Biotechnologie, Laval, Canada
| |
Collapse
|
35
|
Kumar R, Ng S, Engwerda C. The Role of IL-10 in Malaria: A Double Edged Sword. Front Immunol 2019; 10:229. [PMID: 30809232 PMCID: PMC6379449 DOI: 10.3389/fimmu.2019.00229] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/28/2019] [Indexed: 12/11/2022] Open
Abstract
IL-10 produced by CD4+ T cells suppresses inflammation by inhibiting T cell functions and the upstream activities of antigen presenting cells (APCs). IL-10 was first identified in Th2 cells, but has since been described in IFNγ-producing Tbet+ Th1, FoxP3+ CD4+ regulatory T (Treg) and IL-17-producing CD4+ T (Th17) cells, as well as many innate and innate-like immune cell populations. IL-10 production by Th1 cells has emerged as an important mechanism to dampen inflammation in the face of intractable infection, including in African children with malaria. However, although these type I regulatory T (Tr1) cells protect tissue from inflammation, they may also promote disease by suppressing Th1 cell-mediated immunity, thereby allowing infection to persist. IL-10 produced by other immune cells during malaria can also influence disease outcome, but the full impact of this IL-10 production is still unclear. Together, the actions of this potent anti-inflammatory cytokine along with other immunoregulatory mechanisms that emerge following Plasmodium infection represent a potential hurdle for the development of immunity against malaria, whether naturally acquired or vaccine-induced. Recent advances in understanding how IL-10 production is initiated and regulated have revealed new opportunities for manipulating IL-10 for therapeutic advantage. In this review, we will summarize our current knowledge about IL-10 production during malaria and discuss its impact on disease outcome. We will highlight recent advances in our understanding about how IL-10 production by specific immune cell subsets is regulated and consider how this knowledge may be used in drug delivery and vaccination strategies to help eliminate malaria.
Collapse
Affiliation(s)
- Rajiv Kumar
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India.,Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Susanna Ng
- Immunology and Infection Lab, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Christian Engwerda
- Immunology and Infection Lab, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
36
|
Cherrier DE, Serafini N, Di Santo JP. Innate Lymphoid Cell Development: A T Cell Perspective. Immunity 2019; 48:1091-1103. [PMID: 29924975 DOI: 10.1016/j.immuni.2018.05.010] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 05/15/2018] [Accepted: 05/25/2018] [Indexed: 02/08/2023]
Abstract
Innate lymphoid cells (ILCs) and natural killer (NK) cells have garnered considerable interest due to their unique functional properties in immune defense and tissue homeostasis. Our current understanding of how these cells develop has been greatly facilitated by knowledge of T cell biology. Models of T cell differentiation provided the basis for a conceptual classification of these innate effectors and inspired a scheme of their activation and regulation. In this review, we discuss NK cell and ILC development from a "T cell standpoint" in an attempt to extend the analogy between adaptive T cells and their innate ILC and NK cell counterparts.
Collapse
Affiliation(s)
- Dylan E Cherrier
- Innate Immunity Unit, Institut Pasteur, Paris 75015, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1223, Paris 75015, France; Université Paris Diderot, Paris 75013, France
| | - Nicolas Serafini
- Innate Immunity Unit, Institut Pasteur, Paris 75015, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1223, Paris 75015, France
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, Paris 75015, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1223, Paris 75015, France.
| |
Collapse
|
37
|
Bunn PT, Montes de Oca M, de Labastida Rivera F, Kumar R, Ng SS, Edwards CL, Faleiro RJ, Sheel M, Amante FH, Frame TCM, Muller W, Haque A, Uzonna JE, Hill GR, Engwerda CR. Distinct Roles for CD4+ Foxp3+ Regulatory T Cells and IL-10–Mediated Immunoregulatory Mechanisms during Experimental Visceral Leishmaniasis Caused by Leishmania donovani. THE JOURNAL OF IMMUNOLOGY 2018; 201:3362-3372. [DOI: 10.4049/jimmunol.1701582] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 09/25/2018] [Indexed: 12/19/2022]
|
38
|
Burrack KS, Huggins MA, Taras E, Dougherty P, Henzler CM, Yang R, Alter S, Jeng EK, Wong HC, Felices M, Cichocki F, Miller JS, Hart GT, Johnson AJ, Jameson SC, Hamilton SE. Interleukin-15 Complex Treatment Protects Mice from Cerebral Malaria by Inducing Interleukin-10-Producing Natural Killer Cells. Immunity 2018; 48:760-772.e4. [PMID: 29625893 DOI: 10.1016/j.immuni.2018.03.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 11/27/2017] [Accepted: 03/05/2018] [Indexed: 12/21/2022]
Abstract
Cerebral malaria is a deadly complication of Plasmodium infection and involves blood brain barrier (BBB) disruption following infiltration of white blood cells. During experimental cerebral malaria (ECM), mice inoculated with Plasmodium berghei ANKA-infected red blood cells develop a fatal CM-like disease caused by CD8+ T cell-mediated pathology. We found that treatment with interleukin-15 complex (IL-15C) prevented ECM, whereas IL-2C treatment had no effect. IL-15C-expanded natural killer (NK) cells were necessary and sufficient for protection against ECM. IL-15C treatment also decreased CD8+ T cell activation in the brain and prevented BBB breakdown without influencing parasite load. IL-15C induced NK cells to express IL-10, which was required for IL-15C-mediated protection against ECM. Finally, we show that ALT-803, a modified human IL-15C, mediates similar induction of IL-10 in NK cells and protection against ECM. These data identify a regulatory role for cytokine-stimulated NK cells in the prevention of a pathogenic immune response.
Collapse
Affiliation(s)
- Kristina S Burrack
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55414, USA
| | - Matthew A Huggins
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Emily Taras
- Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Philip Dougherty
- Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Christine M Henzler
- Supercomputing Institute for Advanced Computational Research, University of Minnesota, Minneapolis, MN 55414, USA
| | - Rendong Yang
- Supercomputing Institute for Advanced Computational Research, University of Minnesota, Minneapolis, MN 55414, USA
| | - Sarah Alter
- Altor BioScience Corporation, Miramar, FL 33025, USA
| | - Emily K Jeng
- Altor BioScience Corporation, Miramar, FL 33025, USA
| | - Hing C Wong
- Altor BioScience Corporation, Miramar, FL 33025, USA
| | - Martin Felices
- Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Frank Cichocki
- Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Jeffrey S Miller
- Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Geoffrey T Hart
- Center for Immunology, Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Stephen C Jameson
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55414, USA
| | - Sara E Hamilton
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55414, USA.
| |
Collapse
|
39
|
Moyo D, Beattie L, Andrews PS, Moore JWJ, Timmis J, Sawtell A, Hoehme S, Sampson AT, Kaye PM. Macrophage Transactivation for Chemokine Production Identified as a Negative Regulator of Granulomatous Inflammation Using Agent-Based Modeling. Front Immunol 2018; 9:637. [PMID: 29636754 PMCID: PMC5880939 DOI: 10.3389/fimmu.2018.00637] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 03/14/2018] [Indexed: 01/22/2023] Open
Abstract
Cellular activation in trans by interferons, cytokines, and chemokines is a commonly recognized mechanism to amplify immune effector function and limit pathogen spread. However, an optimal host response also requires that collateral damage associated with inflammation is limited. This may be particularly so in the case of granulomatous inflammation, where an excessive number and/or excessively florid granulomas can have significant pathological consequences. Here, we have combined transcriptomics, agent-based modeling, and in vivo experimental approaches to study constraints on hepatic granuloma formation in a murine model of experimental leishmaniasis. We demonstrate that chemokine production by non-infected Kupffer cells in the Leishmania donovani-infected liver promotes competition with infected KCs for available iNKT cells, ultimately inhibiting the extent of granulomatous inflammation. We propose trans-activation for chemokine production as a novel broadly applicable mechanism that may operate early in infection to limit excessive focal inflammation.
Collapse
Affiliation(s)
- Daniel Moyo
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, United Kingdom.,Department of Computer Science, University of York, York, United Kingdom
| | - Lynette Beattie
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, United Kingdom
| | - Paul S Andrews
- Department of Electronics, University of York, York, United Kingdom.,SimOmics Ltd., York, United Kingdom
| | - John W J Moore
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, United Kingdom
| | - Jon Timmis
- Department of Electronics, University of York, York, United Kingdom.,SimOmics Ltd., York, United Kingdom
| | - Amy Sawtell
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, United Kingdom
| | - Stefan Hoehme
- Institute for Computer Science, University of Leipzig, Leipzig, Germany
| | - Adam T Sampson
- Division of Computing and Mathematics, Abertay University, Dundee, United Kingdom
| | - Paul M Kaye
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, United Kingdom
| |
Collapse
|
40
|
Messlinger H, Sebald H, Heger L, Dudziak D, Bogdan C, Schleicher U. Monocyte-Derived Signals Activate Human Natural Killer Cells in Response to Leishmania Parasites. Front Immunol 2018; 9:24. [PMID: 29472914 PMCID: PMC5810259 DOI: 10.3389/fimmu.2018.00024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 01/04/2018] [Indexed: 12/21/2022] Open
Abstract
Activated natural killer (NK) cells release interferon (IFN)-γ, which is crucial for the control of intracellular pathogens such as Leishmania. In contrast to experimental murine leishmaniasis, the human NK cell response to Leishmania is still poorly characterized. Here, we investigated the interaction of human blood NK cells with promastigotes of different Leishmania species (Leishmania major, Leishmania mexicana, Leishmania infantum, and Leishmania donovani). When peripheral blood mononuclear cells or purified NK cells and monocytes (all derived from healthy blood donors from Germany without a history of leishmaniasis) were exposed to promastigotes, NK cells showed increased surface expression of the activation marker CD69. The extent of this effect varied depending on the Leishmania species; differences between dermotropic and viscerotropic L. infantum strains were not observed. Upregulation of CD69 required direct contact between monocytes and Leishmania and was partly inhibitable by anti-interleukin (IL)-18. Unexpectedly, IL-18 was undetectable in most of the supernatants (SNs) of monocyte/parasite cocultures. Confocal fluorescence microscopy of non-permeabilized cells revealed that Leishmania-infected monocytes trans-presented IL-18 to NK cells. Native, but not heat-treated SNs of monocyte/Leishmania cocultures also induced CD69 on NK cells, indicating the involvement of a soluble heat-labile factor other than IL-18. A role for the NK cell-activating cytokines IL-1β, IL-2, IL-12, IL-15, IL-21, and IFN-α/β was excluded. The increase of CD69 was not paralleled by NK cell IFN-γ production or enhanced cytotoxicity. However, prior exposure of NK cells to Leishmania parasites synergistically increased their IFN-γ release in response to IL-12, which was dependent on endogenous IL-18. CD1c+ dendritic cells were identified as possible source of Leishmania-induced IL-12. Finally, we observed that direct contact between Leishmania and NK cells reduced the expression of CD56 mRNA and protein on NK cells. We conclude that Leishmania activate NK cells via trans-presentation of IL-18 by monocytes and by a monocyte-derived soluble factor. IL-12 is needed to elicit the IFN-γ-response of NK cells, which is likely to be an important component of the innate control of the parasite.
Collapse
Affiliation(s)
- Helena Messlinger
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Heidi Sebald
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Lukas Heger
- Laboratory of DC Biology, Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Diana Dudziak
- Laboratory of DC Biology, Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Ulrike Schleicher
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
41
|
Wilkaniec A, Gąssowska-Dobrowolska M, Strawski M, Adamczyk A, Czapski GA. Inhibition of cyclin-dependent kinase 5 affects early neuroinflammatory signalling in murine model of amyloid beta toxicity. J Neuroinflammation 2018; 15:1. [PMID: 29301548 PMCID: PMC5753486 DOI: 10.1186/s12974-017-1027-y] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/07/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Cyclin-dependent kinase 5 (Cdk5) belongs to the family of proline-directed serine/threonine kinases and plays a critical role in neuronal differentiation, migration, synaptogenesis, plasticity, neurotransmission and apoptosis. The deregulation of Cdk5 activity was observed in post mortem analysis of brain tissue of Alzheimer's disease (AD) patients, suggesting the involvement of Cdk5 in the pathomechanism of this neurodegenerative disease. However, our recent study demonstrated the important function of Cdk5 in regulating inflammatory reaction. METHODS Since the role of Cdk5 in regulation of inflammatory signalling in AD is unknown, we investigated the involvement of Cdk5 in neuroinflammation induced by single intracerebroventricular (icv) injection of amyloid beta protein (Aβ) oligomers in mouse. The brain tissue was analysed up to 35 days post injection. Roscovitine (intraperitoneal administration) was used as a potent Cdk5 inhibitor. The experiments were also performed on human neuroblastoma SH-SY5Y as well as mouse BV2 cell lines treated with exogenous oligomeric Aβ. RESULTS Our results demonstrated that single injection of Aβ oligomers induces long-lasting activation of microglia and astrocytes in the hippocampus. We observed also profound, early inflammatory response in the mice hippocampus, leading to the significant elevation of pro-inflammatory cytokines expression (e.g. TNF-α, IL-1β, IL-6). Moreover, Aβ oligomers elevated the formation of truncated protein p25 in mouse hippocampus and induced overactivation of Cdk5 in neuronal cells. Importantly, administration of roscovitine reduced the inflammatory processes evoked by Aβ in the hippocampus, leading to the significant decrease of cytokines level. CONCLUSIONS These studies clearly show the involvement of Cdk5 in modulation of brain inflammatory response induced by Aβ and may indicate this kinase as a novel target for pharmacological intervention in AD.
Collapse
Affiliation(s)
- Anna Wilkaniec
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106, Warsaw, Poland
| | - Magdalena Gąssowska-Dobrowolska
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106, Warsaw, Poland
| | - Marcin Strawski
- Laboratory of Electrochemistry, Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093, Warsaw, Poland
| | - Agata Adamczyk
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106, Warsaw, Poland
| | - Grzegorz A Czapski
- Department of Cellular Signalling, Mossakowski Medical Research Centre Polish Academy of Sciences, Pawińskiego 5, 02-106, Warsaw, Poland.
| |
Collapse
|
42
|
Dixit S, Baganizi DR, Sahu R, Dosunmu E, Chaudhari A, Vig K, Pillai SR, Singh SR, Dennis VA. Immunological challenges associated with artificial skin grafts: available solutions and stem cells in future design of synthetic skin. J Biol Eng 2017; 11:49. [PMID: 29255480 PMCID: PMC5729423 DOI: 10.1186/s13036-017-0089-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 11/17/2017] [Indexed: 12/29/2022] Open
Abstract
The repair or replacement of damaged skins is still an important, challenging public health problem. Immune acceptance and long-term survival of skin grafts represent the major problem to overcome in grafting given that in most situations autografts cannot be used. The emergence of artificial skin substitutes provides alternative treatment with the capacity to reduce the dependency on the increasing demand of cadaver skin grafts. Over the years, considerable research efforts have focused on strategies for skin repair or permanent skin graft transplantations. Available skin substitutes include pre- or post-transplantation treatments of donor cells, stem cell-based therapies, and skin equivalents composed of bio-engineered acellular or cellular skin substitutes. However, skin substitutes are still prone to immunological rejection, and as such, there is currently no skin substitute available to overcome this phenomenon. This review focuses on the mechanisms of skin rejection and tolerance induction and outlines in detail current available strategies and alternatives that may allow achieving full-thickness skin replacement and repair.
Collapse
Affiliation(s)
- Saurabh Dixit
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA.,Immunity, Inflammation, and Disease Laboratory, NIH/NIEHS, Durham, 27709 NC USA
| | - Dieudonné R Baganizi
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Rajnish Sahu
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Ejowke Dosunmu
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Atul Chaudhari
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Komal Vig
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Shreekumar R Pillai
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Shree R Singh
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| | - Vida A Dennis
- Center for Nanobiotechnology Research and Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104 USA
| |
Collapse
|
43
|
Dugast E, David G, Oger R, Danger R, Judor JP, Gagne K, Chesneau M, Degauque N, Soulillou JP, Paul P, Picard C, Guerif P, Conchon S, Giral M, Gervois N, Retière C, Brouard S. Broad Impairment of Natural Killer Cells from Operationally Tolerant Kidney Transplanted Patients. Front Immunol 2017; 8:1721. [PMID: 29312288 PMCID: PMC5732263 DOI: 10.3389/fimmu.2017.01721] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 11/21/2017] [Indexed: 01/10/2023] Open
Abstract
The role of natural killer (NK) cells in organ transplantation is controversial. This study aims to decipher their role in kidney transplant tolerance in humans. Previous studies highlighted several modulated genes involved in NK cell biology in blood from spontaneously operationally tolerant patients (TOLs; drug-free kidney-transplanted recipients with stable graft function). We performed a phenotypic, functional, and genetic characterization of NK cells from these patients compared to kidney-transplanted patients with stable graft function under immunosuppression and healthy volunteers (HVs). Both operationally TOLs and stable patients harbored defective expression of the NKp46 activator receptor and lytic molecules perforin and granzyme compared to HVs. Surprisingly, NK cells from operationally TOLs also displayed decreased expression of the CD16 activating marker (in the CD56Dim NK cell subset). This decrease was associated with impairment of their functional capacities upon stimulation, as shown by lower interferon gamma (IFNγ) production and CD107a membranous expression in a reverse antibody-dependent cellular cytotoxicity (ADCC) assay, spontaneous lysis assays, and lower target cell lysis in the 51Cr release assay compared to HVs. Conversely, despite impaired K562 cell lysis in the 51Cr release assay, patients with stable graft function harbored a normal reverse ADCC and even increased amounts of IFNγ+ NK cells in the spontaneous lysis assay. Altogether, the strong impairment of the phenotype and functional cytotoxic capacities of NK cells in operationally TOLs may accord with the establishment of a pro-tolerogenic environment, despite remaining highly activated after transplantation in patients with stable graft function.
Collapse
Affiliation(s)
- Emilie Dugast
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Gaëlle David
- Etablissement Français du sang, Nantes, France.,CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Romain Oger
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Richard Danger
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Jean-Paul Judor
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Katia Gagne
- Etablissement Français du sang, Nantes, France.,CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,LabEx Transplantex, Université de Strasbourg, France
| | - Mélanie Chesneau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Nicolas Degauque
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | | | - Pascale Paul
- Nephrology Dialysis Renal Transplantation Center, Assistance Publique des Hôpitaux de Marseille, Hospital de la Conception, UMR 1076, Vascular Research Center of Marseille, INSERM, Aix-Marseille University, Marseille, France
| | - Christophe Picard
- Établissement Français du Sang Alpes Méditerranée, Marseille, France.,ADES UMR 7268, CNRS, EFS, Aix-Marseille Université, Marseille, France
| | - Pierrick Guerif
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,CIC Biotherapy, CHU Nantes, Nantes, France
| | - Sophie Conchon
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Magali Giral
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,CIC Biotherapy, CHU Nantes, Nantes, France
| | - Nadine Gervois
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Christelle Retière
- Etablissement Français du sang, Nantes, France.,CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Sophie Brouard
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| |
Collapse
|
44
|
Kesiraju S, Ch UMR, Paritala P, Reddy S, Reddy VS, Sahariah S. Unexplained post-renal transplant tolerance: a case report. Immunol Res 2017; 64:791-4. [PMID: 26690794 DOI: 10.1007/s12026-015-8766-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Achievement of an immunosuppression-free condition defined as clinical operational tolerance is an ideal goal. We hereby report a case of clinical operational tolerance in a patient whose allograft is functioning normally, without immunosuppression, for more than 3 years. The patient withdrew from immunosuppression in 2011 following which his serum creatinine was 1.34 mg/dl and proteinuria was 178 mg/24 h. Flowcytometric studies showed an elevated number of B lymphocytes and NK cells. IL-10 cytokine levels had increased, whereas those of IFN-γ decreased, suggesting that both B lymphocytes and NK cells, with their immunoregulatory function, contribute to the maintenance of long-term graft function. Consequently, further studies in understanding the interactions of NK cells and B lymphocytes may give us a better insight into the underlying mechanisms that underpin organ tolerance.
Collapse
Affiliation(s)
- Sailaja Kesiraju
- Department of Immunology and Biochemistry, Transimmun-Transplantation Immunology and Research Center, Somajiguda, Hyderabad, 500082, India.
| | - Uma Maheswara Rao Ch
- Kidney Transplant Unit, Mahavir Hospital and Research Center, AC Guards, Hyderabad, India
| | - Purna Paritala
- Department of Nephrology and Transplantation, Krishna Institute of Medical Sciences, Hyderabad, India
| | - Sreedhar Reddy
- Department of Nephrology and Transplantation, Krishna Institute of Medical Sciences, Hyderabad, India
| | - V S Reddy
- Department of Nephrology and Transplantation, Krishna Institute of Medical Sciences, Hyderabad, India
| | - S Sahariah
- Department of Immunology and Biochemistry, Transimmun-Transplantation Immunology and Research Center, Somajiguda, Hyderabad, 500082, India
- Kidney Transplant Unit, Mahavir Hospital and Research Center, AC Guards, Hyderabad, India
- Department of Nephrology and Transplantation, Krishna Institute of Medical Sciences, Hyderabad, India
| |
Collapse
|
45
|
Qiu Y, Jiang Z, Hu S, Wang L, Ma X, Yang X. Lactobacillus plantarum Enhanced IL-22 Production in Natural Killer (NK) Cells That Protect the Integrity of Intestinal Epithelial Cell Barrier Damaged by Enterotoxigenic Escherichia coli. Int J Mol Sci 2017; 18:E2409. [PMID: 29137183 PMCID: PMC5713377 DOI: 10.3390/ijms18112409] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/08/2017] [Accepted: 11/08/2017] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL)-22-producing Natural Killer (NK) cells protect the gut epithelial cell barrier from pathogens. A strain of probiotics, Lactobacillus plantarum (L. plantarum, LP), was previously found by our laboratory to significantly improve the mucosal barrier integrity and function of the small intestine in pigs. However, it was unclear whether LP benefited the intestinal mucosal barrier via interactions with the intestinal NK cells. The present study, therefore, was focused on the therapeutic effect of NK cells that were stimulated by LP on attenuating enterotoxigenic Escherichia coli (ETEC)-induced the damage to the integrity of the epithelial cell barrier. The results showed that LP can efficiently increase protein levels of the natural cytotoxicity receptor (NCR) family, and the expression levels of IL-22 mRNA and protein in NK cells. Transfer of NK cells stimulated by LP conferred protection against ETEC K88-induced intestinal epithelial barrier damage in NCM460 cells. We found that NK cells stimulated by LP could partially offset the reduction in NCM460 cell monolayers transepithelial electrical resistance (TEER) caused by ETEC K88, and increase ZO-1 and occludin mRNA and protein expressions by ETEC K88-infected NCM460 cells. Furthermore, adding NK cells that were stimulated by LP to ETEC K88-infected NCM460cells, IL-22R1, p-Stat3, and p-Tyk2 expression by NCM460 cells was increased. Mechanistic experiment showed that NK cells stimulated by LP lost the function of maintaining TEER of NCM460 cells challenged with ETEC K88, when polyclonal anti-IL-22 antibody was used to block IL-22 production. Collectively, our results suggested that LP stimulation of NK could enhance IL-22 production, which might be able to provide defense against ETEC-induced damage to the integrity of intestinal epithelial barrier.
Collapse
Affiliation(s)
- Yueqin Qiu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China.
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou 510640, China.
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangzhou 510640, China.
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China.
- Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China.
| | - Zongyong Jiang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China.
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou 510640, China.
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangzhou 510640, China.
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China.
- Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China.
| | - Shenglan Hu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China.
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou 510640, China.
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangzhou 510640, China.
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China.
- Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China.
| | - Li Wang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China.
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou 510640, China.
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangzhou 510640, China.
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China.
- Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China.
| | - Xianyong Ma
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China.
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou 510640, China.
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangzhou 510640, China.
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China.
- Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China.
| | - Xuefen Yang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China.
- State Key Laboratory of Livestock and Poultry Breeding, Guangzhou 510640, China.
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Guangzhou 510640, China.
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China.
- Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China.
| |
Collapse
|
46
|
Kumar R, Chauhan SB, Ng SS, Sundar S, Engwerda CR. Immune Checkpoint Targets for Host-Directed Therapy to Prevent and Treat Leishmaniasis. Front Immunol 2017; 8:1492. [PMID: 29167671 PMCID: PMC5682306 DOI: 10.3389/fimmu.2017.01492] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/23/2017] [Indexed: 12/15/2022] Open
Abstract
Leishmaniasis encompasses a group of diseases caused by protozoan parasites belonging to the genus Leishmania. These diseases range from life threatening visceral forms to self-healing cutaneous lesions, and each disease manifestations can progress to complications involving dissemination of parasites to skin or mucosal tissue. A feature of leishmaniasis is the key role host immune responses play in disease outcome. T cells are critical for controlling parasite growth. However, they can also contribute to disease onset and progression. For example, potent regulatory T cell responses can develop that suppress antiparasitic immunity. Alternatively, hyperactivated CD4+ or CD8+ T cells can be generated that cause damage to host tissues. There is no licensed human vaccine and drug treatment options are often limited and problematic. Hence, there is an urgent need for new strategies to improve the efficacy of current vaccine candidates and/or enhance both antiparasitic drug effectiveness and subsequent immunity in treated individuals. Here, we describe our current understanding about host immune responses contributing to disease protection and progression in the various forms of leishmaniasis. We also discuss how this knowledge may be used to develop new strategies for host-directed immune therapy to prevent or treat leishmaniasis. Given the major advances made in immune therapy in the cancer and autoimmune fields in recent years, there are significant opportunities to ride on the back of these successes in the infectious disease domain. Conversely, the rapid progress in our understanding about host immune responses during leishmaniasis is also providing opportunities to develop novel immunotherapy strategies that could have broad applications in diseases characterized by inflammation or immune dysfunction.
Collapse
Affiliation(s)
- Rajiv Kumar
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Shashi Bhushan Chauhan
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Susanna S. Ng
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | | |
Collapse
|
47
|
IL-33 promotes IL-10 production in macrophages: a role for IL-33 in macrophage foam cell formation. Exp Mol Med 2017; 49:e388. [PMID: 29099095 PMCID: PMC5704190 DOI: 10.1038/emm.2017.183] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 04/27/2017] [Accepted: 05/02/2017] [Indexed: 12/16/2022] Open
Abstract
We evaluated the role of IL-10- in IL-33-mediated cholesterol reduction in macrophage-derived foam cells (MFCs) and the mechanism by which IL-33 upregulates IL-10. Serum IL-33 and IL-10 levels in coronary artery disease patients were measured. The effects of IL-33 on intra-MFC cholesterol level, IL-10, ABCA1 and CD36 expression, ERK 1/2, Sp1, STAT3 and STAT4 activation, and IL-10 promoter activity were determined. Core sequences were identified using bioinformatic analysis and site-specific mutagenesis. The serum IL-33 levels positively correlated with those of IL-10. IL-33 decreased cellular cholesterol level and upregulated IL-10 and ABCA1 but had no effect on CD36 expression. siRNA-IL-10 partially abolished cellular cholesterol reduction and ABCA1 elevation by IL-33 but did not reverse the decreased CD36 levels. IL-33 increased IL-10 mRNA production but had little effect on its stability. IL-33 induced ERK 1/2 phosphorylation and increased the luciferase expression driven by the IL-10 promoter, with the highest extent within the −2000 to −1752 bp segment of the 5′-flank of the transcription start site; these effects were counteracted by U0126. IL-33 activated Sp1, STAT3 and STAT4, but only the STAT3 binding site was predicted in the above segment. Site-directed mutagenesis of the predicted STAT3-binding sites (CTGCTTCCTGGCAGCAGAA→CTGCCTGGCAGCAGAA) reduced luciferase activity, and a STAT3 inhibitor blocked the regulatory effects of IL-33 on IL-10 expression. Chromatin immunoprecipitation (CHIP) confirmed the STAT3-binding sequences within the −1997 to −1700 and −1091 to −811 bp locus regions. IL-33 increased IL-10 expression in MFCs via activating ERK 1/2 and STAT3, which subsequently promoted IL-10 transcription and thus contributed to the beneficial effects of IL-33 on MFCs.
Collapse
|
48
|
Chauhan P, Shukla D, Chattopadhyay D, Saha B. Redundant and regulatory roles for Toll-like receptors in Leishmania infection. Clin Exp Immunol 2017; 190:167-186. [PMID: 28708252 PMCID: PMC5629438 DOI: 10.1111/cei.13014] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2017] [Indexed: 01/07/2023] Open
Abstract
Toll-like receptors (TLRs) are germline-encoded, non-clonal innate immune receptors, which are often the first receptors to recognize the molecular patterns on pathogens. Therefore, the immune response initiated by TLRs has far-reaching consequences on the outcome of an infection. As soon as the cell surface TLRs and other receptors recognize a pathogen, the pathogen is phagocytosed. Inclusion of TLRs in the phagosome results in quicker phagosomal maturation and stronger adaptive immune response, as TLRs influence co-stimulatory molecule expression and determinant selection by major histocompatibility complex (MHC) class II and MHC class I for cross-presentation. The signals delivered by the TCR-peptide-MHC complex and co-stimulatory molecules are indispensable for optimal T cell activation. In addition, the cytokines induced by TLRs can skew the differentiation of activated T cells to different effector T cell subsets. However, the potential of TLRs to influence adaptive immune response into different patterns is severely restricted by multiple factors: gross specificity for the molecular patterns, lack of receptor rearrangements, sharing of limited number of adaptors that assemble signalling complexes and redundancy in ligand recognition. These features of apparent redundancy and regulation in the functioning of TLRs characterize them as important and probable contributory factors in the resistance or susceptibility to an infection.
Collapse
Affiliation(s)
- P. Chauhan
- Pathogenesis and Cellular Response Division, National Centre for Cell ScienceGaneshkhind, PuneIndia
| | - D. Shukla
- Pathogenesis and Cellular Response Division, National Centre for Cell ScienceGaneshkhind, PuneIndia
| | | | - B. Saha
- National Institute of Traditional MedicineBelagaviIndia
| |
Collapse
|
49
|
Dai YC, Zhong J, Xu JF. Regulatory B cells in infectious disease (Review). Mol Med Rep 2017; 16:3-10. [PMID: 28534949 PMCID: PMC5482109 DOI: 10.3892/mmr.2017.6605] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 03/22/2017] [Indexed: 01/06/2023] Open
Abstract
Regulatory B cells (Bregs) are a subset of B cells, which reportedly exert significant immunomodulatory effects through the production of interleukin (IL)‑10, IL‑35 and transforming growth factor‑β. Over the last decade, studies have indicated that Bregs function in autoimmune and allergic diseases through antigen‑specific and non‑specific immunoregulatory mechanisms. However, only a limited number of reviews have focused on the role of Bregs during infection, particularly their functions in intracellular infections. The present review discusses the role of Bregs in infectious diseases in animal models and human studies, and provides an overview of the immunoregulatory mechanisms used by Bregs.
Collapse
Affiliation(s)
- You-Chao Dai
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan, Guangdong 523808, P.R. China
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical College, Dongguan, Guangdong 523808, P.R. China
| | - Jixin Zhong
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jun-Fa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan, Guangdong 523808, P.R. China
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical College, Dongguan, Guangdong 523808, P.R. China
| |
Collapse
|
50
|
Schuetz C, Lee KM, Scott R, Kojima L, Washburn L, Liu L, Liu WH, Tector H, Lei J, Yeh H, Kim JI, Markmann JF. Regulatory B Cell-Dependent Islet Transplant Tolerance Is Also Natural Killer Cell Dependent. Am J Transplant 2017; 17:1656-1662. [PMID: 28296255 PMCID: PMC5444975 DOI: 10.1111/ajt.14265] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 02/06/2017] [Accepted: 02/22/2017] [Indexed: 01/25/2023]
Abstract
Immunologic tolerance to solid organ and islet cell grafts has been achieved in various rodent models by using antibodies directed at CD45RB and Tim-1. We have shown that this form of tolerance depends on regulatory B cells (Bregs). To elucidate further the mechanism by which Bregs induce tolerance, we investigated the requirement of natural killer (NK) and NKT cells in this model. To do so, hyperglycemic B6, μMT, Beige, or CD1d-/- mice received BALB/c islet grafts and treatment with the tolerance-inducing regimen consisting of anti-CD45RB and anti-TIM1. B6 mice depleted of both NK and NKT cells by anti-NK1.1 antibody and mice deficient in NK activity (Beige) did not develop tolerance after dual-antibody treatment. In contrast, transplant tolerance induction was successful in CD1d-/- recipients (deficient in NKT cells), indicating that NK, but not NKT, cells are essential in B cell-dependent tolerance. In addition, reconstitution of Beige host with NK cells restored the ability to induce transplant tolerance with dual-antibody treatment. Transfer of tolerance by B cells from tolerant mice was also dependent on host Nk1.1+ cells. In conclusion, these results show that regulatory function of B cells is dependent on NK cells in this model of transplantation tolerance.
Collapse
Affiliation(s)
- C Schuetz
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - K M Lee
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - R Scott
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - L Kojima
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - L Washburn
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - L Liu
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - W-H Liu
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - H Tector
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - J Lei
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - H Yeh
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - J I Kim
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - J F Markmann
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Boston, MA
| |
Collapse
|