1
|
Mukherjee D, Satyavolu S, Cioffi S, Thomas A, Li Y, Nayak L. Protocols for circulating neutrophil depletion in neonatal C57Bl/6 mice. J Leukoc Biol 2025; 117:qiaf005. [PMID: 39821323 DOI: 10.1093/jleuko/qiaf005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/12/2024] [Accepted: 01/15/2025] [Indexed: 01/19/2025] Open
Abstract
Murine neonatal neutrophil depletion strategies have problems achieving deep neutrophil clearance and accurate residual neutrophil fraction detection. An isotype switch method can achieve profound neutrophil clearance using a combination of anti-Ly6G and antirat κ Ig light chain antibodies in adult C57Bl/6 mice, proven by extra- and intracellular Ly6G detection by flow cytometry. We adapted this technique to neonatal mice, testing four neutrophil depletion strategies in the peripheral circulation, bone marrow, and spleen. Four protocols were tested: P3 Ly6G and P1-3 Ly6G (anti-Ly6G on postnatal days [P] 3 and 1-3, respectively), and P3 Dual and P1-3 Dual (anti-Ly6G and antirat κ Ig light chain on P3 and P1-3, respectively). Intracellular and extracellular Ly6G presence was detected using flow cytometry. Isotype control antibodies were used as controls. P1-3 Dual protocol achieved significantly better neutrophil depletion than the P1-3 Ly6G or P3 Ly6G protocols (97% vs 74% and 97% vs 50%, respectively) in the peripheral circulation. The P3 Dual protocol alone was enough to achieve significantly better neutrophil clearance (93%) than any of the Ly6G alone protocols. The Ly6G alone protocols led to near-total elimination of extracellular Ly6G. However, there was a significant presence of intracellular Ly6G in the CD45+ cell population, evading detection by extracellular Ly6G antibody-based detection methods. P3 protocols perform better than P1-3 protocols for bone marrow and splenic neutrophil clearance. Thus, the P3 Dual protocol might be the most effective and ethical protocol to induce profound neutrophil depletion in neonatal mice, an alternative to daily anti-Ly6G injections.
Collapse
Affiliation(s)
- Devashis Mukherjee
- Department of Pediatrics, Case Western Reserve University School of Medicine, 11100 Euclid Ave, Cleveland, OH 44106, United States
- Rainbow Babies and Children's Hospital, 11100 Euclid Ave, Cleveland, OH 44106, United States
| | - Sriram Satyavolu
- The Ohio State University College of Medicine, 1645 Neil Ave, Columbus, OH 43210, United States
| | - Sarah Cioffi
- Department of Pediatrics, Case Western Reserve University School of Medicine, 11100 Euclid Ave, Cleveland, OH 44106, United States
| | - Asha Thomas
- Case Comprehensive Cancer Center, 2103 Cornell Rd, Cleveland, OH 44106, United States
| | - Yuexin Li
- Case Comprehensive Cancer Center, 2103 Cornell Rd, Cleveland, OH 44106, United States
| | - Lalitha Nayak
- Case Comprehensive Cancer Center, 2103 Cornell Rd, Cleveland, OH 44106, United States
- Division of Hematology-Oncology, Department of Medicine, University Hospitals Cleveland Medical Center, 11100 Euclid Ave, Cleveland, OH 44106, United States
| |
Collapse
|
2
|
Yuan Z, Luozhong S, Li R, Gu W, Chen Y, Bhashyam D, Lai R, Jiang S. Transient Macrophage Depletion Circumvents Scavenging and Redirects Biodistribution of mRNA-Lipid Nanoparticles. ACS NANO 2025; 19:14422-14433. [PMID: 40167042 DOI: 10.1021/acsnano.5c02001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The mononuclear phagocytic system is recognized as a major scavenger of mRNA-lipid nanoparticles (LNPs), clearing and redirecting these particles away from their intended targets and thus diminishing their delivery efficacy. Understanding the mechanism by which mRNA-LNPs interact with phagocytes and how this interaction affects the mRNA transfection is critical to enhancing the delivery of mRNA. In this study, we temporarily depleted both circulating and resident macrophages (MF) and evaluated the transfection efficiency and biodistribution of mRNA-LNPs. We first demonstrated the enhanced liver expression using two liver-tropic formulations and the significant improvement of the in vivo gene editing efficiency of CRISPR-Cas9 in the Ai14 mouse model after MF depletion, providing a versatile strategy for enhanced mRNA delivery to the liver regardless of the formulation employed. We then extended our investigations to lung-tropic and lymphoid-tropic LNP formulations and discovered that MF depletion abolishes the targeting capacities of these non-liver-tropic formulations, providing insights into the organ targeting of LNPs. Finally, we screened and compared various clinically relevant MF depletion methods, providing the translation potential of this method on enhanced hepatic delivery of mRNA.
Collapse
Affiliation(s)
- Zhefan Yuan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Sijin Luozhong
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Ruoxin Li
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Wenchao Gu
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Yu Chen
- Department of Materials Science and Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Dani Bhashyam
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Rachel Lai
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Shaoyi Jiang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
3
|
Chen J, Bai Y, He X, Xiao W, Chen L, Wong YK, Wang C, Gao P, Cheng G, Xu L, Yang C, Liao F, Han G, Sun J, Xu C, Wang J. The spatiotemporal transcriptional profiling of murine brain during cerebral malaria progression and after artemisinin treatment. Nat Commun 2025; 16:1540. [PMID: 39934099 PMCID: PMC11814382 DOI: 10.1038/s41467-024-52223-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 08/28/2024] [Indexed: 02/13/2025] Open
Abstract
Cerebral malaria (CM) is a severe encephalopathy caused by Plasmodium parasite infection, resulting in thousands of annual deaths and neuro-cognitive sequelae even after anti-malarial drugs treatment. Despite efforts to dissect the mechanism, the cellular transcriptomic reprogramming within the spatial context remains elusive. Here, we constructed single-cell and spatial transcriptome atlases of experimental CM (ECM) male murine brain tissues with or without artesunate (ART) treatment. We identified activated inflammatory endothelial cells during ECM, characterized by a disrupted blood-brain barrier, increased antigen presentation, and leukocyte adhesion. We also observed that inflammatory microglia enhance antigen presentation pathway such as MHC-I to CD8+ cytotoxic T cells. The latter underwent an inflammatory state transition with up-regulated cytokine expression and cytotoxic activity. Multi-omics analysis revealed that the activated interferon-gamma response of injured neurons during ECM and persisted after ART treatment. Overall, our research provides valuable resources for understanding malaria parasite-host interaction mechanisms and adjuvant therapy development.
Collapse
Affiliation(s)
- Jiayun Chen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China
| | - Yunmeng Bai
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Xueling He
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Wei Xiao
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China
- Department of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Lina Chen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yin Kwan Wong
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Chen Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Peng Gao
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, Henan, China
| | - Guangqing Cheng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Liting Xu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chuanbin Yang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Fulong Liao
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Guang Han
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, Henan, China
| | - Jichao Sun
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| | - Chengchao Xu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| | - Jigang Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China.
- Department of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, Henan, China.
| |
Collapse
|
4
|
Meng Q, Seto F, Totsu T, Miyashita T, Wu S, Bougaki M, Ushio M, Hiruma T, Trapnell BC, Uchida K. Lung immune incompetency after mild peritoneal sepsis and its partial restoration by type 1 interferon: a mouse model study. Intensive Care Med Exp 2024; 12:119. [PMID: 39707074 DOI: 10.1186/s40635-024-00707-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Sepsis is commonly associated with acute respiratory distress syndrome (ARDS). Although the exaggerated inflammation may damage intact lung tissues, a percentage of patients with ARDS are reportedly immunocompromised, with worse outcomes. Herein, using a murine sepsis model, time-course immune reprogramming after sepsis was evaluated to explore whether the host is immunocompromised. Leukocyte kinetics in the lung tissue were evaluated in a male C57/BL6 mouse model of mild peritoneal sepsis induced by cecal ligation and puncture, with the survival rate exceeds 90%. Lung immune reactivity was evaluated by intratracheal instillation of lipopolysaccharide (LPS; 30 µg). Furthermore, the effect of interferon (IFN)-β in vivo and ex vivo was evaluated. RESULTS Four days after sepsis, the lung water content remained high, even among mice in clinical recovery. While monocytes and neutrophils gradually accumulated in the lung interstitium, the inflammatory cytokine/chemokine expression levels in the lungs continued to decline. Intratracheal LPS instillation induced more leukocyte trafficking and protein leakage into the alveoli in the septic lung, indicating more severe lung injury. However, LPS stimulation-associated mRNA expression of tnf, il6, ccl2, and cxcl1 was suppressed. Intra-alveolar expression of tumor necrosis factor (TNF)-α, interleukin (IL)-6, monocyte chemoattractant protein (MCP)-1, and keratinocyte-derived cytokine (KC) was also suppressed. Monocytes isolated from the lung tissue showed an impaired response in il6, ccl2, and cxcl1 to LPS. Systemic IFN-β restored the above impaired regulator function of monocytes, as did coculturing these cells from lung tissue with IFN-β. CONCLUSIONS Histologically accelerated inflammation and paradoxically suppressed immunological regulator signaling were observed in the early recovery phase of sepsis. This observation may provide a model for the immunologically irresponsive state that occurs in some patients with sepsis. Systemic IFN-β partly restored the post-septic immunocompromised state, indicating its therapeutic potential for the immunosuppressive state seen in some patients with sepsis/ARDS.
Collapse
Affiliation(s)
- Qiuming Meng
- Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Fumiko Seto
- Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Tokie Totsu
- Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Tomoyuki Miyashita
- Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Songfei Wu
- Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Masahiko Bougaki
- Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Michiko Ushio
- Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Takahiro Hiruma
- Department of Emergency and Intensive Care Medicine, Department of Emergency and Critical Care Medicine, Fukushima Medical University, Southern Tohoku Research Institute for Neuroscience, Southern Tohoku General Hospital, Fukushima, Japan
| | - Bruce C Trapnell
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kanji Uchida
- Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
5
|
Owen AR, Farias A, Levins AM, Wang Z, Higham SL, Mack M, Tregoning JS, Johansson C. Exposure to bacterial PAMPs before RSV infection exacerbates innate inflammation and disease via IL-1α and TNF-α. Mucosal Immunol 2024; 17:1184-1198. [PMID: 39127259 PMCID: PMC11631774 DOI: 10.1016/j.mucimm.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/17/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
Respiratory syncytial virus (RSV) can cause severe lower respiratory tract infections. Understanding why some individuals get more serious disease may help with diagnosis and treatment. One possible risk factor underlying severe disease is bacterial exposure before RSV infection. Bacterial exposure has been associated with increased respiratory viral-induced disease severity but the mechanism remains unknown. Respiratory bacterial infections or exposure to their pathogen associated molecular patterns (PAMPs) trigger innate immune inflammation, characterised by neutrophil and inflammatory monocyte recruitment and the production of inflammatory cytokines. We hypothesise that these changes to the lung environment alter the immune response and disease severity during subsequent RSV infection. To test this, we intranasally exposed mice to LPS, LTA or Acinetobacter baumannii (an airway bacterial pathogen) before RSV infection and observed an early induction of disease, measured by weight loss, at days 1-3 after infection. Neutrophils or inflammatory monocytes were not responsible for driving this exacerbated weight loss. Instead, exacerbated disease was associated with increased IL-1α and TNF-α, which orchestrated the recruitment of innate immune cells into the lung. This study shows that exposure to bacterial PAMPs prior to RSV infection increases the expression of IL-1α and TNF-α, which dysregulate the immune response resulting in exacerbated disease.
Collapse
Affiliation(s)
- Amber R Owen
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Ana Farias
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Anne-Marie Levins
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, United Kingdom
| | - Ziyin Wang
- Department of Infectious Disease, Imperial College London, United Kingdom
| | - Sophie L Higham
- Department of Infectious Disease, Imperial College London, United Kingdom
| | - Matthias Mack
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - John S Tregoning
- Department of Infectious Disease, Imperial College London, United Kingdom
| | - Cecilia Johansson
- Respiratory Infections, National Heart and Lung Institute, Imperial College London, United Kingdom.
| |
Collapse
|
6
|
Mukherjee D, Satyavolu S, Cioffi S, Thomas A, Li Y, Nayak L. Protocols for circulating neutrophil depletion in neonatal C57Bl/6 mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592835. [PMID: 39282419 PMCID: PMC11398316 DOI: 10.1101/2024.05.06.592835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Murine neonatal neutrophil depletion strategies have problems achieving deep neutrophil clearance and accurate residual neutrophil fraction detection. An isotype switch method can achieve profound neutrophil clearance using a combination of anti-Ly6G and anti-rat κ Ig light chain antibodies in adult C57Bl/6 mice, proven by extra- and intracellular Ly6G detection by flow cytometry. We adapted this technique to neonatal mice, testing four neutrophil depletion strategies in the peripheral circulation, bone marrow, and spleen. Four protocols were tested: P3 Ly6G and P1-3 Ly6G (anti-Ly6G on postnatal days (P) 3 and 1-3 respectively), and P3 Dual and P1-3 Dual (anti-Ly6G and anti-rat κ Ig light chain on P3 and P1-3 respectively). Intracellular and extracellular Ly6G presence was detected using flow cytometry. Isotype control antibodies were used as controls. P1-3 Dual protocol achieved significantly better neutrophil depletion than the P1-3 Ly6G or P3 Ly6G protocols (97% vs. 74% and 97% vs. 50%, respectively) in the peripheral circulation. The P3 Dual protocol alone was enough to achieve significantly better neutrophil clearance (93%) than any of the Ly6G alone protocols. The Ly6G alone protocols led to near-total elimination of extracellular Ly6G. However, there was a significant presence of intracellular Ly6G in the CD45+ cell population, evading detection by extracellular Ly6G antibody-based detection methods. P3 protocols perform better than P1-3 protocols for bone marrow and splenic neutrophil clearance. Thus, the P3 Dual protocol might be the most effective and ethical protocol to induce profound neutrophil depletion in neonatal mice, an alternative to daily anti-Ly6G injections. Summary Sentence Dual antibody-based neutrophil depletion effectively induces circulating neutrophil clearance in neonatal mice.
Collapse
|
7
|
Sellau J, Hansen CS, Gálvez RI, Linnemann L, Honecker B, Lotter H. Immunological clues to sex differences in parasitic diseases. Trends Parasitol 2024; 40:1029-1041. [PMID: 39379261 DOI: 10.1016/j.pt.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/23/2024] [Accepted: 09/12/2024] [Indexed: 10/10/2024]
Abstract
The effect of sex on the prevalence and severity of parasitic diseases is an emerging area of research. Several factors underlie sex-based differences, including sociocultural influences that affect exposure to parasites, and physiological disparities linked to biological sex. Hence, human studies must be interpreted cautiously; however, studies conducted under controlled laboratory conditions are important to validate findings in humans. Such research can more effectively elucidate the role of sex-determining physiological factors (particularly their impact on immune responses), as well as the role of sex-specific differences in resistance to, or severity of, parasitic diseases. This review focuses on the overarching impact of biological sex variables on immunity. Both human and rodent experimental data are discussed, with a focus on selected protozoan and helminth infections.
Collapse
Affiliation(s)
- Julie Sellau
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | | | - Lara Linnemann
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Barbara Honecker
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Hanna Lotter
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
| |
Collapse
|
8
|
Zhu Y, Zhou L, Mo L, Hong C, Pan L, Lin J, Qi Y, Tan S, Qian M, Hu T, Zhao Y, Qiu H, Lin P, Ma X, Yang Q. Plasmodium yoelii Infection Enhances the Expansion of Myeloid-Derived Suppressor Cells via JAK/STAT3 Pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:170-186. [PMID: 38819229 DOI: 10.4049/jimmunol.2300541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 05/07/2024] [Indexed: 06/01/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs), the negative immune regulators, have been demonstrated to be involved in immune responses to a variety of pathological conditions, such as tumors, chronic inflammation, and infectious diseases. However, the roles and mechanisms underlying the expansion of MDSCs in malaria remain unclear. In this study, the phenotypic and functional characteristics of splenic MDSCs during Plasmodium yoelii NSM infection are described. Furthermore, we provide compelling evidence that the sera from P. yoelii-infected C57BL/6 mice containing excess IL-6 and granulocyte-macrophage colony-stimulating factor promote the accumulation of MDSCs by inducing Bcl2 expression. Serum-induced MDSCs exert more potent suppressive effects on T cell responses than control MDSCs within both in vivo P. yoelii infection and in vitro serum-treated bone marrow cells experiments. Serum treatment increases the MDSC inhibitory effect, which is dependent on Arg1 expression. Moreover, mechanistic studies reveal that the serum effects are mediated by JAK/STAT3 signaling. By inhibiting STAT3 phosphorylation with the JAK inhibitor JSI-124, effects of serum on MDSCs are almost eliminated. In vivo depletion of MDSCs with anti-Gr-1 or 5-fluorouracil significantly reduces the parasitemia and promotes Th1 immune response in P. yoelii-infected C57BL/6 mice by upregulating IFN-γ expression. In summary, this study indicates that P. yoelii infection facilitates the accumulation and function of MDSCs by upregulating the expression of Bcl2 and Arg1 via JAK/STAT3 signaling pathway in vivo and in vitro. Manipulating the JAK/STAT3 signaling pathway or depleting MDSCs could be promising therapeutic interventions to treat malaria.
Collapse
Affiliation(s)
- Yiqiang Zhu
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Lu Zhou
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Lengshan Mo
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Cansheng Hong
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Lingxia Pan
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jie Lin
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yanwei Qi
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Simin Tan
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Manhongtian Qian
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Tengfei Hu
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yi Zhao
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Huaina Qiu
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Peibin Lin
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Xiancai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, China
| | - Quan Yang
- Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
9
|
Wassmer SC, de Koning-Ward TF, Grau GER, Pai S. Unravelling mysteries at the perivascular space: a new rationale for cerebral malaria pathogenesis. Trends Parasitol 2024; 40:28-44. [PMID: 38065791 PMCID: PMC11072469 DOI: 10.1016/j.pt.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 01/06/2024]
Abstract
Cerebral malaria (CM) is a severe neurological complication caused by Plasmodium falciparum parasites; it is characterized by the sequestration of infected red blood cells within the cerebral microvasculature. New findings, combined with a better understanding of the central nervous system (CNS) barriers, have provided greater insight into the players and events involved in CM, including site-specific T cell responses in the human brain. Here, we review the updated roles of innate and adaptive immune responses in CM, with a focus on the role of the perivascular macrophage-endothelium unit in antigen presentation, in the vascular and perivascular compartments. We suggest that these events may be pivotal in the development of CM.
Collapse
Affiliation(s)
- Samuel C Wassmer
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK.
| | - Tania F de Koning-Ward
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia; Institute of Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Victoria, Australia
| | - Georges E R Grau
- Vascular Immunology Unit, Discipline of Pathology, School of Medical Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Saparna Pai
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia.
| |
Collapse
|
10
|
Orchanian SB, Lodoen MB. Monocytes as primary defenders against Toxoplasma gondii infection. Trends Parasitol 2023; 39:837-849. [PMID: 37633758 DOI: 10.1016/j.pt.2023.07.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/28/2023]
Abstract
Monocytes are recruited from the bone marrow to sites of infection where they release cytokines and chemokines, function in antimicrobial immunity, and differentiate into macrophages and dendritic cells to control infection. Although many studies have focused on monocyte-derived macrophages and dendritic cells, recent work has examined the unique roles of monocytes during infection to promote immune defense. We focus on the effector functions of monocytes during infection with the parasite Toxoplasma gondii, and discuss the signals that mobilize monocytes to sites of infection, their production of inflammatory cytokines and antimicrobial mediators, their ability to shape the adaptive immune response, and their immunoregulatory functions. Insights from other infections, including Plasmodium and Listeria are also included for comparison and context.
Collapse
Affiliation(s)
- Stephanie B Orchanian
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California, USA; Institute for Immunology, University of California Irvine, Irvine, California, USA
| | - Melissa B Lodoen
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California, USA; Institute for Immunology, University of California Irvine, Irvine, California, USA.
| |
Collapse
|
11
|
Freire-Antunes L, Ornellas-Garcia U, Rangel-Ferreira MV, Ribeiro-Almeida ML, de Sousa CHG, Carvalho LJDM, Daniel-Ribeiro CT, Ribeiro-Gomes FL. Increased Neutrophil Percentage and Neutrophil-T Cell Ratio Precedes Clinical Onset of Experimental Cerebral Malaria. Int J Mol Sci 2023; 24:11332. [PMID: 37511092 PMCID: PMC10379066 DOI: 10.3390/ijms241411332] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/07/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Newly emerging data suggest that several neutrophil defense mechanisms may play a role in both aggravating and protecting against malaria. These exciting findings suggest that the balance of these cells in the host body may have an impact on the pathogenesis of malaria. To fully understand the role of neutrophils in severe forms of malaria, such as cerebral malaria (CM), it is critical to gain a comprehensive understanding of their behavior and functions. This study investigated the dynamics of neutrophil and T cell responses in C57BL/6 and BALB/c mice infected with Plasmodium berghei ANKA, murine models of experimental cerebral malaria (ECM) and non-cerebral experimental malaria, respectively. The results demonstrated an increase in neutrophil percentage and neutrophil-T cell ratios in the spleen and blood before the development of clinical signs of ECM, which is a phenomenon not observed in the non-susceptible model of cerebral malaria. Furthermore, despite the development of distinct forms of malaria in the two strains of infected animals, parasitemia levels showed equivalent increases throughout the infection period evaluated. These findings suggest that the neutrophil percentage and neutrophil-T cell ratios may be valuable predictive tools for assessing the dynamics and composition of immune responses involved in the determinism of ECM development, thus contributing to the advancing of our understanding of its pathogenesis.
Collapse
Affiliation(s)
- Lucas Freire-Antunes
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Rio de Janeiro 21041-250, Brazil
| | - Uyla Ornellas-Garcia
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Rio de Janeiro 21041-250, Brazil
| | - Marcos Vinicius Rangel-Ferreira
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Rio de Janeiro 21041-250, Brazil
| | - Mônica Lucas Ribeiro-Almeida
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Rio de Janeiro 21041-250, Brazil
| | - Carina Heusner Gonçalves de Sousa
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Rio de Janeiro 21041-250, Brazil
| | - Leonardo José de Moura Carvalho
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Rio de Janeiro 21041-250, Brazil
| | - Cláudio Tadeu Daniel-Ribeiro
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Rio de Janeiro 21041-250, Brazil
| | - Flávia Lima Ribeiro-Gomes
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Rio de Janeiro 21041-250, Brazil
| |
Collapse
|
12
|
Shin AE, Tesfagiorgis Y, Larsen F, Derouet M, Zeng PYF, Good HJ, Zhang L, Rubinstein MR, Han YW, Kerfoot SM, Nichols AC, Hayakawa Y, Howlett CJ, Wang TC, Asfaha S. F4/80 +Ly6C high Macrophages Lead to Cell Plasticity and Cancer Initiation in Colitis. Gastroenterology 2023; 164:593-609.e13. [PMID: 36634827 PMCID: PMC10038892 DOI: 10.1053/j.gastro.2023.01.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 12/15/2022] [Accepted: 01/03/2023] [Indexed: 01/14/2023]
Abstract
BACKGROUND & AIMS Colorectal cancer is a leading cause of cancer death, and a major risk factor is chronic inflammation. Despite the link between colitis and cancer, the mechanism by which inflammation leads to colorectal cancer is not well understood. METHODS To investigate whether different forms of inflammation pose the same risk of cancer, we compared several murine models of colitis (dextran sodium sulfate [DSS], 2,4,6-trinitrobenzene sulfonic acid, 4-ethoxylmethylene-2-phenyloxazol-5-one, Citrobacter rodentium, Fusobacterium nucleatum, and doxorubicin) with respect to their ability to lead to colonic tumorigenesis. We attempted to correlate the severity of colitis and inflammatory profile with the risk of tumorigenesis in both azoxymethane-dependent and Dclk1/APCfl/fl murine models of colitis-associated cancer. RESULTS DSS colitis reproducibly led to colonic tumors in both mouse models of colitis-associated cancer. In contrast, all other forms of colitis did not lead to cancer. When compared with the colitis not associated with tumorigenesis, DSS colitis was characterized by significantly increased CD11b+F4/80+Ly6Chigh macrophages and CD11b+Ly6G+ neutrophils. Interestingly, depletion of the CD11b+F4/80+Ly6Chigh macrophages inhibited tumorigenesis, whereas depletion of CD11b+Ly6G+ neutrophils had no effect on tumorigenesis. Furthermore, the macrophage-derived cytokines interleukin-1β, tumor necrosis factor-α, and interleukin-6 were significantly increased in DSS colitis and promoted stemness of Dclk1+ tuft cells that serve as the cellular origin of cancer. CONCLUSIONS We have identified CD11b+F4/80+Ly6Chigh macrophages as key mediators of cancer initiation in colitis-associated cancer. Development of new therapies that target these cells may provide an effective preventative strategy for colitis-associated cancer.
Collapse
Affiliation(s)
- Alice E Shin
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Department of Medicine, Western University, London, Ontario, Canada
| | - Yodit Tesfagiorgis
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Frederikke Larsen
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Department of Medicine, Western University, London, Ontario, Canada
| | - Mathieu Derouet
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Department of Medicine, Western University, London, Ontario, Canada
| | - Peter Y F Zeng
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Department of Otolaryngology and Head and Neck Surgery, Western University, London, Ontario, Canada
| | - Hayley J Good
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Department of Medicine, Western University, London, Ontario, Canada
| | - Liyue Zhang
- Department of Medicine, Western University, London, Ontario, Canada
| | - Mara R Rubinstein
- Section of Oral, Diagnostic and Rehabilitation Sciences, College of Dental Medicine, Columbia University Irving Medical Center, New York, New York
| | - Yiping W Han
- Section of Oral, Diagnostic and Rehabilitation Sciences, College of Dental Medicine, Columbia University Irving Medical Center, New York, New York; Departments of Microbiology & Immunology and Medicine (Medical Sciences), Columbia University Irving Medical Center, New York, New York
| | - Steven M Kerfoot
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Anthony C Nichols
- Department of Otolaryngology and Head and Neck Surgery, Western University, London, Ontario, Canada
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Christopher J Howlett
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Timothy C Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Samuel Asfaha
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Department of Medicine, Western University, London, Ontario, Canada.
| |
Collapse
|
13
|
Gopinath A, Mackie PM, Phan LT, Mirabel R, Smith AR, Miller E, Franks S, Syed O, Riaz T, Law BK, Urs N, Khoshbouei H. Who Knew? Dopamine Transporter Activity Is Critical in Innate and Adaptive Immune Responses. Cells 2023; 12:cells12020269. [PMID: 36672204 PMCID: PMC9857305 DOI: 10.3390/cells12020269] [Citation(s) in RCA: 11] [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: 11/29/2022] [Revised: 12/22/2022] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
The dopamine transporter (DAT) regulates the dimension and duration of dopamine transmission. DAT expression, its trafficking, protein-protein interactions, and its activity are conventionally studied in the CNS and within the context of neurological diseases such as Parkinson's Diseases and neuropsychiatric diseases such as drug addiction, attention deficit hyperactivity and autism. However, DAT is also expressed at the plasma membrane of peripheral immune cells such as monocytes, macrophages, T-cells, and B-cells. DAT activity via an autocrine/paracrine signaling loop regulates macrophage responses to immune stimulation. In a recent study, we identified an immunosuppressive function for DAT, where blockade of DAT activity enhanced LPS-mediated production of IL-6, TNF-α, and mitochondrial superoxide levels, demonstrating that DAT activity regulates macrophage immune responses. In the current study, we tested the hypothesis that in the DAT knockout mice, innate and adaptive immunity are perturbed. We found that genetic deletion of DAT (DAT-/-) results in an exaggerated baseline inflammatory phenotype in peripheral circulating myeloid cells. In peritoneal macrophages obtained from DAT-/- mice, we identified increased MHC-II expression and exaggerated phagocytic response to LPS-induced immune stimulation, suppressed T-cell populations at baseline and following systemic endotoxemia and exaggerated memory B cell expansion. In DAT-/- mice, norepinephrine and dopamine levels are increased in spleen and thymus, but not in circulating serum. These findings in conjunction with spleen hypoplasia, increased splenic myeloid cells, and elevated MHC-II expression, in DAT-/- mice further support a critical role for DAT activity in peripheral immunity. While the current study is only focused on identifying the role of DAT in peripheral immunity, our data point to a much broader implication of DAT activity than previously thought. This study is dedicated to the memory of Dr. Marc Caron who has left an indelible mark in the dopamine transporter field.
Collapse
Affiliation(s)
- Adithya Gopinath
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
- Correspondence: (A.G.); (H.K.)
| | - Phillip M. Mackie
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Leah T. Phan
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Rosa Mirabel
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32611, USA
| | - Aidan R. Smith
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Emily Miller
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Stephen Franks
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Ohee Syed
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Tabish Riaz
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Brian K. Law
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32611, USA
| | - Nikhil Urs
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32611, USA
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
- Correspondence: (A.G.); (H.K.)
| |
Collapse
|
14
|
Leonardo L, Kenangalem E, Poespoprodjo JR, Noviyanti R, Price RN, Anstey NM, Minigo G, Kho S. Increased circulating myeloid-derived suppressor cells in vivax malaria and severe falciparum malaria. Malar J 2022; 21:255. [PMID: 36068577 PMCID: PMC9446641 DOI: 10.1186/s12936-022-04268-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 08/16/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Circulating myeloid-derived-suppressor-cells (MDSC) with immunosuppressive function are increased in human experimental Plasmodium falciparum infection, but have not been studied in clinical malaria. METHODS Using flow-cytometry, circulating polymorphonuclear-MDSC were evaluated in cryopreserved samples from patients with uncomplicated Plasmodium vivax (n = 8) and uncomplicated (n = 4) and severe (n = 16) falciparum malaria from Papua, Indonesia. RESULTS The absolute number of circulating polymorphonuclear-MDSC were significantly elevated in severe falciparum malaria patients compared to controls (n = 10). Polymorphonuclear-MDSC levels in uncomplicated vivax malaria were also elevated to levels comparable to that seen in severe falciparum malaria. CONCLUSION Control of expansion of immunosuppressive MDSC may be important for development of effective immune responses in falciparum and vivax malaria.
Collapse
Affiliation(s)
- Leo Leonardo
- Papuan Health and Community Development Foundation, Timika, Papua, Indonesia
| | - Enny Kenangalem
- Papuan Health and Community Development Foundation, Timika, Papua, Indonesia
| | - Jeanne R Poespoprodjo
- Papuan Health and Community Development Foundation, Timika, Papua, Indonesia
- Department of Pediatrics, University of Gadjah Mada, Yogyakarta, Indonesia
| | | | - Ric N Price
- Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
- Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX37LJ, UK
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nicholas M Anstey
- Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
| | - Gabriela Minigo
- Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
- College of Health and Human Sciences, Charles Darwin University, Darwin, NT, Australia
| | - Steven Kho
- Papuan Health and Community Development Foundation, Timika, Papua, Indonesia.
- Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia.
| |
Collapse
|
15
|
Babatunde KA, Adenuga OF. Neutrophils in malaria: A double-edged sword role. Front Immunol 2022; 13:922377. [PMID: 35967409 PMCID: PMC9367684 DOI: 10.3389/fimmu.2022.922377] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 07/01/2022] [Indexed: 11/16/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in human peripheral blood. They form the first line of defense against invading foreign pathogens and might play a crucial role in malaria. According to World Health Organization (WHO), malaria is a globally significant disease caused by protozoan parasites from the Plasmodium genus, and it's responsible for 627,000 deaths in 2020. Neutrophils participate in the defense response against the malaria parasite via phagocytosis and reactive oxygen species (ROS) production. Neutrophils might also be involved in the pathogenesis of malaria by the release of toxic granules and the release of neutrophil extracellular traps (NETs). Intriguingly, malaria parasites inhibit the anti-microbial function of neutrophils, thus making malaria patients more susceptible to secondary opportunistic Salmonella infections. In this review, we will provide a summary of the role of neutrophils during malaria infection, some contradicting mouse model neutrophil data and neutrophil-related mechanisms involved in malaria patients' susceptibility to bacterial infection.
Collapse
Affiliation(s)
- Kehinde Adebayo Babatunde
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, United States
| | | |
Collapse
|
16
|
Silva Pereira S, De Niz M, Serre K, Ouarné M, Coelho JE, Franco CA, Figueiredo L. Immunopathology and Trypanosoma congolense parasite sequestration cause acute cerebral trypanosomiasis. eLife 2022; 11:77440. [PMID: 35787830 PMCID: PMC9307270 DOI: 10.7554/elife.77440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 07/04/2022] [Indexed: 11/15/2022] Open
Abstract
Trypanosoma congolense causes a syndrome of variable severity in animals in Africa. Cerebral trypanosomiasis is a severe form, but the mechanism underlying this severity remains unknown. We developed a mouse model of acute cerebral trypanosomiasis and characterized the cellular, behavioral, and physiological consequences of this infection. We show large parasite sequestration in the brain vasculature for long periods of time (up to 8 hr) and extensive neuropathology that associate with ICAM1-mediated recruitment and accumulation of T cells in the brain parenchyma. Antibody-mediated ICAM1 blocking and lymphocyte absence reduce parasite sequestration in the brain and prevent the onset of cerebral trypanosomiasis. Here, we establish a mouse model of acute cerebral trypanosomiasis and we propose a mechanism whereby parasite sequestration, host ICAM1, and CD4+ T cells play a pivotal role.
Collapse
Affiliation(s)
- Sara Silva Pereira
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Mariana De Niz
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Karine Serre
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Marie Ouarné
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Joana E Coelho
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Cláudio A Franco
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| | - Luisa Figueiredo
- Instituto de Medicina Molecular, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
17
|
An update on cerebral malaria for therapeutic intervention. Mol Biol Rep 2022; 49:10579-10591. [PMID: 35670928 DOI: 10.1007/s11033-022-07625-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 05/20/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Cerebral malaria is often pronounced as a major life-threatening neurological complication of Plasmodium falciparum infection. The complex pathogenic landscape of the parasite and the associated neurological complications are still not elucidated properly. The growing concerns of drugresistant parasite strains along with the failure of anti-malarial drugs to subdue post-recovery neuro-cognitive dysfunctions in cerebral malaria patients have called for a demand to explore novel biomarkers and therapeutic avenues. Due course of the brain infection journey of the parasite, events such as sequestration of infected RBCs, cytoadherence, inflammation, endothelial activation, and blood-brain barrier disruption are considered critical. METHODS In this review, we briefly summarize the diverse pathogenesis of the brain-invading parasite associated with loss of the blood-brain barrier integrity. In addition, we also discuss proteomics, transcriptomics, and bioinformatics strategies to identify an array of new biomarkers and drug candidates. CONCLUSION A proper understanding of the parasite biology and mechanism of barrier disruption coupled with emerging state-of-art therapeutic approaches could be helpful to tackle cerebral malaria.
Collapse
|
18
|
IL-6 dependent expansion of inflammatory MDSCs (CD11b+ Gr-1+) promote Th-17 mediated immune response during experimental cerebral malaria. Cytokine 2022; 155:155910. [PMID: 35594680 DOI: 10.1016/j.cyto.2022.155910] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 12/18/2022]
Abstract
Myeloid derived suppressor cells (MDSCs) are a group of heterogeneous cell populations that can suppress T cell responses. Various aspects of MDSCs in regulating immune responses in several cancer and infectious diseases have been reported till date. But the role and regulation of MDSCs have not been systematically studied in the context of malaria. This study depicts the phenotypic and functional characteristics of splenic MDSCs and how they regulate Th-17 mediated immune response during Experimental Cerebral Malaria (ECM). Flow cytometric analysis reveals that MDSCs in the spleen and bone marrow expand at 8 dpi during ECM. Among subtypes of MDSCs, PMN-MDSCs show significant expansion in the spleen but M-MDSCs remain unaltered. Functional analysis of sorted MDSCs from spleens of Plasmodium berghei ANKA (PbA) infected mice shows suppressive nature of these cells and high production of Nitric oxide (NO). Besides, MDSCs were also found to express various inflammatory markers during ECM suggesting the M1 type phenotype of these cells. In-vivo depletion of MDSCs by the use of Anti Gr-1 increases mice survival but doesn't significantly alter the parasitemia. Previously, it has been reported that Treg/Th-17 balance in the spleen is skewed towards Th-17 during ECM. Depletion of MDSCs was found to regulate Th-17 percentages to homeostatic levels and subvert various inflammatory changes in the spleen. Among different factors, IL-6 was found to play an important role in the expansion of MDSCs and expression of inflammatory markers on MDSCs in a STAT3-dependent manner. These findings provide a unique insight into the role of IL-6 in the expansion of the MDSC population which causes inflammatory changes and increased Th-17 responses during ECM.
Collapse
|
19
|
Wu X, You D, Cui J, Yang L, Lin L, Chen Y, Xu C, Lian G, Wan J. Reduced Neutrophil Extracellular Trap Formation During Ischemia Reperfusion Injury in C3 KO Mice: C3 Requirement for NETs Release. Front Immunol 2022; 13:781273. [PMID: 35250972 PMCID: PMC8889019 DOI: 10.3389/fimmu.2022.781273] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/20/2022] [Indexed: 12/27/2022] Open
Abstract
Complement C3 plays a prominent role in inflammatory processes, and its increase exacerbates ischemia reperfusion injury (IRI)-induced acute kidney injury (AKI). Infiltrated neutrophils can be stimulated to form neutrophil extracellular traps (NETs), leading to renal injury. However, the relationship between the increase of C3 and the release of NETs in AKI was not clear. Here we found that IRI in the mouse kidney leads to increased neutrophils infiltration and NET formation. Furthermore, neutrophils depletion by anti-Ly6G IgG (1A8) did not reduce C3 activation but reduced kidney injury and inflammation, indicating a link between neutrophils infiltration and renal tissue damage. Pretreatment with 1A8 suppressed ischemia-induced NET formation, proving that extracellular traps (ETs) in renal tissue were mainly derived from neutrophils. Renal ischemia injury also leads to increased expression of C3. Moreover, C3 KO mice (C3 KO) with IRI exhibited attenuated kidney damage and decreased neutrophils and NETs. In vitro, C3a primed neutrophils to form NETs, reflected by amorphous extracellular DNA structures that colocalized with CitH3 and MPO. These data reveal that C3 deficiency can ameliorate AKI by reducing the infiltration of neutrophils and the formation of NETs. Targeting C3 activation may be a new therapeutic strategy for alleviating the necroinflammation of NETs in AKI.
Collapse
Affiliation(s)
- Xiaoting Wu
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Danyu You
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jiong Cui
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Liyan Yang
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Liyu Lin
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yi Chen
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Changsheng Xu
- Fujian Hypertension Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guili Lian
- Fujian Hypertension Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jianxin Wan
- Department of Nephrology, Blood Purification Research Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
20
|
Tamadaho RSE, Ritter M, Wiszniewsky A, Arndts K, Mack M, Hoerauf A, Layland LE. Infection-Derived Monocytic MDSCs Require TGF-β to Suppress Filarial-Specific IFN-γ But Not IL-13 Release by Filarial-Specific CD4+ T Cells In Vitro. FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2021.707100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Lymphatic filariasis (LF) remains a major health problem with severe economic repercussions in endemic communities of Sub-saharan Africa, South-East Asia and South America. The rodent-specific nematode Litomosoides sigmodontis (Ls) is used to study the immunomodulatory potential of filariae and research has elucidated pathways involving regulatory T cells (Tregs), IL-10 producing cells and alternatively activated macrophages (AAMs) and that CD4+ T cells play a paramount role during infection. Myeloid-derived suppressor cells (MDSCs) have been identified and characterised in man in cancer and other pathologies. The hallmark of MDSC populations is the suppression of T and B cell responses using various mechanisms, which are mostly specific to the pathology or setting. However, until now, it remains unclear whether they play a role in filarial-specific responses. We report here that monocytic MDSCs (Mo-MDSCs, CD11b+Ly6C+Ly6G-) and polymorphonuclear MDSCs (PMN-MDSCs, CD11b+Ly6Cint/loLy6G+) expanded in the thoracic cavity (TC, the site of infection) and correlated positively with filarial life-stages in Ls-infected BALB/c mice. In vitro, only infection-derived Mo-MDSCs showed a suppressive nature by preventing IL-13 and IFN-γ secretion from filarial-specific CD4+ T cells upon co-culture with soluble worm extract. This suppression was not mediated by IL-10, IL-6 or TNF-α, and did not require cell-contact, nitric oxide (NO), IL-4/IL-5 signalling pathways or CCR2. Interestingly, neutralizing TGF-β significantly rescued IFN-γ but not IL-13 production by filarial-specific CD4+ T cells. In comparison to naive cells, PCR array data showed an overall down-regulation of inflammatory pathways in both infection-derived Mo-MDSCs and PMN-MDSCs. In conclusion, these primary data sets show activity and expansion of MDSCs during Ls infection adding this regulatory cell type to the complex milieu of host responses during chronic helminth infections.
Collapse
|
21
|
Pollenus E, Gouwy M, Van den Steen PE. Neutrophils in malaria: the good, the bad or the ugly? Parasite Immunol 2022; 44:e12912. [PMID: 35175636 DOI: 10.1111/pim.12912] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/09/2022] [Accepted: 02/13/2022] [Indexed: 11/30/2022]
Abstract
Neutrophils are the most abundant circulating leukocytes in human peripheral blood. They are often the first cells to respond to an invading pathogen and might therefore play an important role in malaria. Malaria is a globally important disease caused by Plasmodium parasites, responsible for more than 400 000 deaths each year. Most of these deaths are caused by complications, including cerebral malaria, severe malarial anemia, placental malaria, renal injury, metabolic problems and malaria-associated acute respiratory distress syndrome. Neutrophils contribute in the immune defense against malaria, through clearance of parasites via phagocytosis, production of reactive oxygen species and release of neutrophil extracellular traps (NETs). However, Plasmodium parasites diminish antibacterial functions of neutrophils, making patients more susceptible to other infections. Neutrophils might also be involved in the development of malaria complications, for example via the release of toxic granules and NETs. However, technical pitfalls in the determination of the roles of neutrophils have caused contradicting results. Further investigations need to consider these pitfalls, in order to elucidate the role of neutrophils in malaria complications.
Collapse
Affiliation(s)
- Emilie Pollenus
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, University of Leuven, Leuven, Belgium
| | - Mieke Gouwy
- Laboratory of Molecular immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, University of Leuven, Leuven, Belgium
| | - Philippe E Van den Steen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, University of Leuven, Leuven, Belgium
| |
Collapse
|
22
|
Scheunemann JF, Reichwald JJ, Korir PJ, Kuehlwein JM, Jenster LM, Hammerschmidt-Kamper C, Lewis MD, Klocke K, Borsche M, Schwendt KE, Soun C, Thiebes S, Limmer A, Engel DR, Mueller AK, Hoerauf A, Hübner MP, Schumak B. Eosinophils Suppress the Migration of T Cells Into the Brain of Plasmodium berghei-Infected Ifnar1-/- Mice and Protect Them From Experimental Cerebral Malaria. Front Immunol 2021; 12:711876. [PMID: 34659202 PMCID: PMC8514736 DOI: 10.3389/fimmu.2021.711876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/13/2021] [Indexed: 11/29/2022] Open
Abstract
Cerebral malaria is a potentially lethal disease, which is caused by excessive inflammatory responses to Plasmodium parasites. Here we use a newly developed transgenic Plasmodium berghei ANKA (PbAAma1OVA) parasite that can be used to study parasite-specific T cell responses. Our present study demonstrates that Ifnar1-/- mice, which lack type I interferon receptor-dependent signaling, are protected from experimental cerebral malaria (ECM) when infected with this novel parasite. Although CD8+ T cell responses generated in the spleen are essential for the development of ECM, we measured comparable parasite-specific cytotoxic T cell responses in ECM-protected Ifnar1-/- mice and wild type mice suffering from ECM. Importantly, CD8+ T cells were increased in the spleens of ECM-protected Ifnar1-/- mice and the blood-brain-barrier remained intact. This was associated with elevated splenic levels of CCL5, a T cell and eosinophil chemotactic chemokine, which was mainly produced by eosinophils, and an increase in eosinophil numbers. Depletion of eosinophils enhanced CD8+ T cell infiltration into the brain and increased ECM induction in PbAAma1OVA-infected Ifnar1-/- mice. However, eosinophil-depletion did not reduce the CD8+ T cell population in the spleen or reduce splenic CCL5 concentrations. Our study demonstrates that eosinophils impact CD8+ T cell migration and proliferation during PbAAma1OVA-infection in Ifnar1-/- mice and thereby are contributing to the protection from ECM.
Collapse
Affiliation(s)
- Johanna F Scheunemann
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Julia J Reichwald
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Patricia Jebett Korir
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Janina M Kuehlwein
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Lea-Marie Jenster
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | | | - Matthew D Lewis
- Parasitology Unit, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Katrin Klocke
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Max Borsche
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Kim E Schwendt
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Camille Soun
- Institute for Experimental Immunology and Imaging, University Hospital Essen, Essen, Germany
| | - Stephanie Thiebes
- Institute for Experimental Immunology and Imaging, University Hospital Essen, Essen, Germany
| | - Andreas Limmer
- Clinic for Anesthesiology and Intensive Care, University Hospital Essen, Essen, Germany
| | - Daniel R Engel
- Institute for Experimental Immunology and Imaging, University Hospital Essen, Essen, Germany
| | - Ann-Kristin Mueller
- Parasitology Unit, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Infection Research (DZIF), Heidelberg, Germany
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany.,German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Marc P Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany.,German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Beatrix Schumak
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
23
|
Xu J, Ganguly A, Zhao J, Ivey M, Lopez R, Osterholzer JJ, Cho CS, Olszewski MA. CCR2 Signaling Promotes Brain Infiltration of Inflammatory Monocytes and Contributes to Neuropathology during Cryptococcal Meningoencephalitis. mBio 2021; 12:e0107621. [PMID: 34311579 PMCID: PMC8406332 DOI: 10.1128/mbio.01076-21] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/25/2021] [Indexed: 12/21/2022] Open
Abstract
Cryptococcal meningoencephalitis (CM) is a leading cause of central nervous system (CNS) infection-related mortality worldwide, with surviving patients often developing neurological deficiencies. While CNS inflammation has been implicated in the pathogenesis of CM, little is known about the relative contribution of the specific inflammatory/immune pathways to CNS pathology versus fungal clearance. Increased cerebrospinal fluid level of C-C chemokine receptor 2 (CCR2) ligand CCL2 is associated with disease deterioration in patients with CM. Using a murine model, we investigated the role of the CCR2 pathway in the development of CNS inflammation and pathology during CM. We found that CCR2-deficient mice exhibited improved 28-day survival and alleviated neurological disease scores despite a brain fungal burden higher than that of the WT mice. Reduced CM pathology in CCR2-deficient mice was accompanied by markedly decreased neuronal cell death around cryptococcal microcysts and restored expression of genes involved in neurotransmission, connectivity, and neuronal cell structure in the brains. Results show that CCR2 axis is the major pathway recruiting CD45hiCD11b+Ly6C+ inflammatory monocyte to the brain and indirectly modulates the accumulation of CD4+ T cells and CD8+ T cells. In particular, CCR2 axis promotes recruitment of interferon gamma (IFN-γ)-producing CD4+ T cells and classical activation of myeloid cells. In this context, CCR2 deletion limits the immune network dysregulation we see in CM and attenuates neuropathology. Thus, the CCR2 axis is a potential target for interventions aimed to limit inflammatory CNS pathology in CM patients. IMPORTANCE Cryptococcal meningoencephalitis (CM) causes nearly 200,000 deaths worldwide each year, and survivors frequently develop long-lasting neurological sequelae. The high rate of mortality and neurologic sequelae in CM patients indicate that antifungal therapies alone are often insufficient to control disease progression. Here, we reveal that CM disease progression in mice is accompanied by inflammatory monocytes infiltration at the periphery of the infected foci that overlap locally perturbed neuronal function and death. Importantly, we identified that CCR2 signaling is a critical pathway driving neuroinflammation, especially inflammatory monocyte recruitment, as well as CNS pathology and mortality in CM mice. Our results imply that targeting the CCR2 pathway may be beneficial as a therapy complementary to antifungal drug treatment, helping to reduce CNS damage and mortality in CM patients.
Collapse
Affiliation(s)
- Jintao Xu
- Research Service, Ann Arbor VA Health System, Department of Veterans Affairs Health System, Ann Arbor, Michigan, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Anutosh Ganguly
- Research Service, Ann Arbor VA Health System, Department of Veterans Affairs Health System, Ann Arbor, Michigan, USA
- Division of Hepatopancreatobiliary and Advanced Gastrointestinal Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Jessica Zhao
- Research Service, Ann Arbor VA Health System, Department of Veterans Affairs Health System, Ann Arbor, Michigan, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Michel Ivey
- Research Service, Ann Arbor VA Health System, Department of Veterans Affairs Health System, Ann Arbor, Michigan, USA
| | - Rafael Lopez
- Research Service, Ann Arbor VA Health System, Department of Veterans Affairs Health System, Ann Arbor, Michigan, USA
| | - John J. Osterholzer
- Research Service, Ann Arbor VA Health System, Department of Veterans Affairs Health System, Ann Arbor, Michigan, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Clifford S. Cho
- Research Service, Ann Arbor VA Health System, Department of Veterans Affairs Health System, Ann Arbor, Michigan, USA
- Division of Hepatopancreatobiliary and Advanced Gastrointestinal Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Michal A. Olszewski
- Research Service, Ann Arbor VA Health System, Department of Veterans Affairs Health System, Ann Arbor, Michigan, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| |
Collapse
|
24
|
Qin J, Lovelace MD, Mitchell AJ, de Koning-Ward T, Grau GE, Pai S. Perivascular macrophages create an intravascular niche for CD8 + T cell localisation prior to the onset of fatal experimental cerebral malaria. Clin Transl Immunology 2021; 10:e1273. [PMID: 33854773 PMCID: PMC8026342 DOI: 10.1002/cti2.1273] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/03/2021] [Accepted: 03/14/2021] [Indexed: 12/12/2022] Open
Abstract
Objectives The immunologic events that build up to the fatal neurological stage of experimental cerebral malaria (ECM) are incompletely understood. Here, we dissect immune cell behaviour occurring in the central nervous system (CNS) when Plasmodium berghei ANKA (PbA)‐infected mice show only minor clinical signs. Methods A 2‐photon intravital microscopy (2P‐IVM) brain imaging model was used to study the spatiotemporal context of early immunological events in situ during ECM. Results Early in the disease course, antigen‐specific CD8+ T cells came in contact and arrested on the endothelium of post‐capillary venules. CD8+ T cells typically adhered adjacent to, or were in the near vicinity of, perivascular macrophages (PVMs) that line post‐capillary venules. Closer examination revealed that CD8+ T cells crawled along the inner vessel wall towards PVMs that lay on the abluminal side of large post‐capillary venules. ‘Activity hotspots’ in large post‐capillary venules were characterised by T‐cell localisation, activated morphology and clustering of PVM, increased abutting of post‐capillary venules by PVM and augmented monocyte accumulation. In the later stages of infection, when mice exhibited neurological signs, intravascular CD8+ T cells increased in number and changed their behaviour, actively crawling along the endothelium and displaying frequent, short‐term interactions with the inner vessel wall at hotspots. Conclusion Our study suggests an active interaction between PVM and CD8+ T cells occurs across the blood–brain barrier (BBB) in early ECM, which may be the initiating event in the inflammatory cascade leading to BBB alteration and neuropathology.
Collapse
Affiliation(s)
| | - Michael D Lovelace
- Applied Neurosciences Program Peter Duncan Neurosciences Research Unit St Vincent's Centre for Applied Medical Research Sydney NSW Australia.,UNSW St Vincent's Clinical School Faculty of Medicine UNSW Sydney Sydney NSW Australia
| | - Andrew J Mitchell
- Materials Characterisation and Fabrication Platform Department of Chemical Engineering University of Melbourne Parkville VIC Australia
| | | | - Georges Er Grau
- Vascular Immunology Unit Discipline of Pathology School of Medical Sciences University of Sydney Camperdown NSW Australia
| | - Saparna Pai
- Centre for Molecular Therapeutics Australian Institute of Tropical Health and Medicine James Cook University Cairns QLD Australia.,Faculty of Medicine and Health University of Sydney Sydney NSW Australia
| |
Collapse
|
25
|
Yinda CK, Port JR, Bushmaker T, Offei Owusu I, Purushotham JN, Avanzato VA, Fischer RJ, Schulz JE, Holbrook MG, Hebner MJ, Rosenke R, Thomas T, Marzi A, Best SM, de Wit E, Shaia C, van Doremalen N, Munster VJ. K18-hACE2 mice develop respiratory disease resembling severe COVID-19. PLoS Pathog 2021; 17:e1009195. [PMID: 33465158 PMCID: PMC7875348 DOI: 10.1371/journal.ppat.1009195] [Citation(s) in RCA: 218] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 02/10/2021] [Accepted: 11/30/2020] [Indexed: 02/06/2023] Open
Abstract
SARS-CoV-2 emerged in late 2019 and resulted in the ongoing COVID-19 pandemic. Several animal models have been rapidly developed that recapitulate the asymptomatic to moderate disease spectrum. Now, there is a direct need for additional small animal models to study the pathogenesis of severe COVID-19 and for fast-tracked medical countermeasure development. Here, we show that transgenic mice expressing the human SARS-CoV-2 receptor (angiotensin-converting enzyme 2 [hACE2]) under a cytokeratin 18 promoter (K18) are susceptible to SARS-CoV-2 and that infection resulted in a dose-dependent lethal disease course. After inoculation with either 104 TCID50 or 105 TCID50, the SARS-CoV-2 infection resulted in rapid weight loss in both groups and uniform lethality in the 105 TCID50 group. High levels of viral RNA shedding were observed from the upper and lower respiratory tract and intermittent shedding was observed from the intestinal tract. Inoculation with SARS-CoV-2 resulted in upper and lower respiratory tract infection with high infectious virus titers in nasal turbinates, trachea and lungs. The observed interstitial pneumonia and pulmonary pathology, with SARS-CoV-2 replication evident in pneumocytes, were similar to that reported in severe cases of COVID-19. SARS-CoV-2 infection resulted in macrophage and lymphocyte infiltration in the lungs and upregulation of Th1 and proinflammatory cytokines/chemokines. Extrapulmonary replication of SARS-CoV-2 was observed in the cerebral cortex and hippocampus of several animals at 7 DPI but not at 3 DPI. The rapid inflammatory response and observed pathology bears resemblance to COVID-19. Additionally, we demonstrate that a mild disease course can be simulated by low dose infection with 102 TCID50 SARS-CoV-2, resulting in minimal clinical manifestation and near uniform survival. Taken together, these data support future application of this model to studies of pathogenesis and medical countermeasure development.
Collapse
Affiliation(s)
- Claude Kwe Yinda
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Julia R. Port
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Trenton Bushmaker
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Irene Offei Owusu
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Jyothi N. Purushotham
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Victoria A. Avanzato
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Robert J. Fischer
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Jonathan E. Schulz
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Myndi G. Holbrook
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Madison J. Hebner
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Rebecca Rosenke
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Tina Thomas
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Andrea Marzi
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Sonja M. Best
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Emmie de Wit
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Carl Shaia
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Neeltje van Doremalen
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Vincent J. Munster
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| |
Collapse
|
26
|
Assis PA, Fernandes Durso D, Chacon Cavalcante F, Zaniratto R, Carvalho-Silva AC, Cunha-Neto E, Golenbock DT, Rodrigues Pinto Ferreira L, Tostes Gazzinelli R. Integrative analysis of microRNA and mRNA expression profiles of monocyte-derived dendritic cells differentiation during experimental cerebral malaria. J Leukoc Biol 2020; 108:1183-1197. [PMID: 32362022 PMCID: PMC11215656 DOI: 10.1002/jlb.1ma0320-731r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/19/2020] [Accepted: 03/23/2020] [Indexed: 12/27/2022] Open
Abstract
Heterogeneity and high plasticity are common features of cells from the mononuclear phagocyte system: monocytes (MOs), macrophages, and dendritic cells (DCs). Upon activation by microbial agents, MO can differentiate into MO-derived DCs (MODCs). In previous work, we have shown that during acute infection with Plasmodium berghei ANKA (PbA), MODCs become, transiently, the main CD11b+ myeloid population in the spleen (SP) and once recruited to the brain play an important role in the development of experimental cerebral malaria (ECM). Here, we isolated 4 cell populations: bone marrow (BM) MOs (BM-MOs) and SP-MOs from uninfected mice; BM inflammatory MOs (BM-iMOs) and SP-MODCs from PbA-infected mice and used a system biology approach to a holistic transcriptomic comparison and provide an interactome analysis by integrating differentially expressed miRNAs (DEMs) and their differentially expressed gene targets (DEGs) data. The Jaccard index (JI) was used for gauging the similarity and diversity among these cell populations. Whereas BM-MOs, BM-iMOs, and SP-MOs presented high similarity of DEGs, SP-MODCs distinguished by showing a greater number of DEGs. Moreover, functional analysis identified an enrichment in canonical pathways, such as DC maturation, neuroinflammation, and IFN signaling. Upstream regulator analysis identified IFNγ as the potential upstream molecule that can explain the observed DEMs-Target DEGs intersections in SP-MODCs. Finally, directed target analysis and in vivo/ex vivo assays indicate that SP-MODCs differentiate in the SP and IFNγ is a main driver of this process.
Collapse
Affiliation(s)
| | - Danielle Fernandes Durso
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | | - Ricardo Zaniratto
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Ana Carolina Carvalho-Silva
- RNA Systems Biology Laboratory (RSBL), Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Douglas Taylor Golenbock
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Ludmila Rodrigues Pinto Ferreira
- RNA Systems Biology Laboratory (RSBL), Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ricardo Tostes Gazzinelli
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Laboratory of Immunopathology, Fundação Oswaldo Cruz - Minas, Belo Horizonte, Minas Gerais, Brazil
- Plataforma de Medicina Translacional, Fundação Oswaldo Cruz/Faculdade de Medicina de Ribeirão Preto, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
27
|
Rodrigues DAS, Prestes EB, Gama AMS, Silva LDS, Pinheiro AAS, Ribeiro JMC, Campos RMP, Pimentel-Coelho PM, De Souza HS, Dicko A, Duffy PE, Fried M, Francischetti IMB, Saraiva EM, Paula-Neto HA, Bozza MT. CXCR4 and MIF are required for neutrophil extracellular trap release triggered by Plasmodium-infected erythrocytes. PLoS Pathog 2020; 16:e1008230. [PMID: 32797076 PMCID: PMC7449500 DOI: 10.1371/journal.ppat.1008230] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 08/26/2020] [Accepted: 06/17/2020] [Indexed: 11/18/2022] Open
Abstract
Neutrophil extracellular traps (NETs) evolved as a unique effector mechanism contributing to resistance against infection that can also promote tissue damage in inflammatory conditions. Malaria infection can trigger NET release, but the mechanisms and consequences of NET formation in this context remain poorly characterized. Here we show that patients suffering from severe malaria had increased amounts of circulating DNA and increased neutrophil elastase (NE) levels in plasma. We used cultured erythrocytes and isolated human neutrophils to show that Plasmodium-infected red blood cells release macrophage migration inhibitory factor (MIF), which in turn caused NET formation by neutrophils in a mechanism dependent on the C-X-C chemokine receptor type 4 (CXCR4). NET production was dependent on histone citrullination by peptidyl arginine deiminase-4 (PAD4) and independent of reactive oxygen species (ROS), myeloperoxidase (MPO) or NE. In vitro, NETs functioned to restrain parasite dissemination in a mechanism dependent on MPO and NE activities. Finally, C57/B6 mice infected with P. berghei ANKA, a well-established model of cerebral malaria, presented high amounts of circulating DNA, while treatment with DNAse increased parasitemia and accelerated mortality, indicating a role for NETs in resistance against Plasmodium infection.
Collapse
Affiliation(s)
- Danielle A. S. Rodrigues
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elisa B. Prestes
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andreza M. S. Gama
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leandro de Souza Silva
- Laboratório de Sinalização Celular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Acácia S. Pinheiro
- Laboratório de Sinalização Celular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jose Marcos C. Ribeiro
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, United States of America
| | - Raquel M. P. Campos
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro
| | - Pedro M. Pimentel-Coelho
- Laboratório de Neurobiologia Celular e Molecular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro
| | - Heitor S. De Souza
- Serviço de Gastroenterologia & Laboratório Multidisciplinar de Pesquisa, Departmento de Medicina Interna, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro and Instituto D’Or para Pesquisa e Educação (IDOR), Rio de Janeiro, Brazil
| | - Alassane Dicko
- Malaria Research & Training Center, Faculty of Medicine, Pharmacy and Dentistry, University of Sciences Techniques and Technologies of Bamako, Bamako, Mali
| | - Patrick E. Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Michal Fried
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ivo M. B. Francischetti
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, United States of America
| | - Elvira M. Saraiva
- Laboratório de Imunobiologia das Leishmanioses, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Heitor A. Paula-Neto
- Laboratório de Alvos Moleculares, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo T. Bozza
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
28
|
Sellau J, Groneberg M, Fehling H, Thye T, Hoenow S, Marggraff C, Weskamm M, Hansen C, Stanelle-Bertram S, Kuehl S, Noll J, Wolf V, Metwally NG, Hagen SH, Dorn C, Wernecke J, Ittrich H, Tannich E, Jacobs T, Bruchhaus I, Altfeld M, Lotter H. Androgens predispose males to monocyte-mediated immunopathology by inducing the expression of leukocyte recruitment factor CXCL1. Nat Commun 2020; 11:3459. [PMID: 32651360 PMCID: PMC7351718 DOI: 10.1038/s41467-020-17260-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 06/18/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatic amebiasis, predominantly occurring in men, is a focal destruction of the liver due to the invading protozoan Entamoeba histolytica. Classical monocytes as well as testosterone are identified to have important functions for the development of hepatic amebiasis in mice, but a link between testosterone and monocytes has not been identified. Here we show that testosterone treatment induces proinflammatory responses in human and mouse classical monocytes. When treated with 5α-dihydrotestosterone, a strong androgen receptor ligand, human classical monocytes increase CXCL1 production in the presence of Entamoeba histolytica antigens. Moreover, plasma testosterone levels of individuals undergoing transgender procedure correlate positively with the TNF and CXCL1 secretion from their cultured peripheral blood mononuclear cells following lipopolysaccharide stimulation. Finally, testosterone substitution of castrated male mice increases the frequency of TNF/CXCL1-producing classical monocytes during hepatic amebiasis, supporting the hypothesis that the effects of androgens may contribute to an increased risk of developing monocyte-mediated pathologies.
Collapse
Affiliation(s)
- Julie Sellau
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Marie Groneberg
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Helena Fehling
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Thorsten Thye
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Stefan Hoenow
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Claudia Marggraff
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Marie Weskamm
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Charlotte Hansen
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Stephanie Stanelle-Bertram
- Department Viral Zoonoses - One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Svenja Kuehl
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Jill Noll
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Vincent Wolf
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Nahla Galal Metwally
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Sven Hendrik Hagen
- Research Department Virus Immunology, Heinrich Pette Institute, Hamburg, Germany
| | | | - Julia Wernecke
- Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Harald Ittrich
- Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Egbert Tannich
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Thomas Jacobs
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Iris Bruchhaus
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Marcus Altfeld
- Research Department Virus Immunology, Heinrich Pette Institute, Hamburg, Germany.,Department of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hannelore Lotter
- Department of Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
| |
Collapse
|
29
|
Sala E, Kuka M. The Suppressive Attitude of Inflammatory Monocytes in Antiviral Antibody Responses. Viral Immunol 2020; 33:327-333. [PMID: 32027238 PMCID: PMC7247028 DOI: 10.1089/vim.2019.0132] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Inflammatory monocytes play important functions in antiviral immune responses, including release of inflammatory cytokines and antigen presentation to T lymphocytes. Depending on the pathological context, these functions might translate into beneficial or detrimental effects in the resolution of the disease. Recent literature has highlighted a role for inflammatory monocytes also in direct suppression of B cell responses. In this review, we will briefly discuss research showing the relationship between inflammatory monocytes and B lymphocytes, its functional consequences on antiviral antibody responses, and possible implications in the design of future vaccination strategies.
Collapse
Affiliation(s)
- Eleonora Sala
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Mirela Kuka
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
30
|
Kuehlwein JM, Borsche M, Korir PJ, Risch F, Mueller A, Hübner MP, Hildner K, Hoerauf A, Dunay IR, Schumak B. Protection of Batf3-deficient mice from experimental cerebral malaria correlates with impaired cytotoxic T-cell responses and immune regulation. Immunology 2020; 159:193-204. [PMID: 31631339 PMCID: PMC6954726 DOI: 10.1111/imm.13137] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 09/30/2019] [Accepted: 10/14/2019] [Indexed: 12/28/2022] Open
Abstract
Excessive inflammatory immune responses during infections with Plasmodium parasites are responsible for severe complications such as cerebral malaria (CM) that can be studied experimentally in mice. Dendritic cells (DCs) activate cytotoxic CD8+ T-cells and initiate immune responses against the parasites. Batf3-/- mice lack a DC subset, which efficiently induces strong CD8 T-cell responses by cross-presentation of exogenous antigens. Here we show that Batf3-/- mice infected with Plasmodium berghei ANKA (PbA) were protected from experimental CM (ECM), characterized by a stable blood-brain barrier (BBB) and significantly less infiltrated peripheral immune cells in the brain. Importantly, the absence of ECM in Batf3-/- mice correlated with attenuated responses of cytotoxic T-cells, as their parasite-specific lytic activity as well as the production of interferon gamma and granzyme B were significantly decreased. Remarkably, spleens of ECM-protected Batf3-/- mice had elevated levels of regulatory immune cells and interleukin 10. Thus, protection from ECM in PbA-infected Batf3-/- mice was associated with the absence of strong CD8+ T-cell activity and induction of immunoregulatory mediators and cells.
Collapse
MESH Headings
- Animals
- Basic-Leucine Zipper Transcription Factors/deficiency
- Basic-Leucine Zipper Transcription Factors/genetics
- Blood-Brain Barrier/immunology
- Blood-Brain Barrier/parasitology
- Brain/immunology
- Brain/metabolism
- Brain/parasitology
- Cells, Cultured
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/parasitology
- Disease Models, Animal
- Female
- Granzymes/immunology
- Granzymes/metabolism
- Host-Parasite Interactions
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Interleukin-10/immunology
- Interleukin-10/metabolism
- Malaria, Cerebral/immunology
- Malaria, Cerebral/metabolism
- Malaria, Cerebral/parasitology
- Malaria, Cerebral/prevention & control
- Mice, Inbred C57BL
- Mice, Knockout
- Plasmodium berghei/immunology
- Plasmodium berghei/pathogenicity
- Repressor Proteins/deficiency
- Repressor Proteins/genetics
- Spleen/immunology
- Spleen/metabolism
- Spleen/parasitology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/parasitology
Collapse
Affiliation(s)
- Janina M. Kuehlwein
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
| | - Max Borsche
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
| | - Patricia J. Korir
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
| | - Frederic Risch
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
| | - Ann‐Kristin Mueller
- Parasitology UnitCentre for Infectious DiseasesHeidelberg University HospitalHeidelbergGermany
- DZIF German Center for Infection ResearchPartner Site HeidelbergHeidelbergGermany
| | - Marc P. Hübner
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
| | - Kai Hildner
- Medical Department 1University Hospital ErlangenErlangenGermany
| | - Achim Hoerauf
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
- DZIF German Center for Infection ResearchPartner Site Bonn‐CologneBonnGermany
| | - Ildiko Rita Dunay
- Institute of Inflammation and NeurodegenerationUniversity of MagdeburgMagdeburgGermany
| | - Beatrix Schumak
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
| |
Collapse
|
31
|
Calvo B, Rubio F, Fernández M, Tranque P. Dissociation of neonatal and adult mice brain for simultaneous analysis of microglia, astrocytes and infiltrating lymphocytes by flow cytometry. IBRO Rep 2020; 8:36-47. [PMID: 32215337 PMCID: PMC7090101 DOI: 10.1016/j.ibror.2019.12.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 12/31/2019] [Indexed: 12/29/2022] Open
Abstract
Recovery of neural cells is higher with 30 % Percoll gradient than 30–70 %. Papain enhances combined extraction of microglia, astrocytes and lymphocytes. Dispase II potentiates papain action only in adult brain. Mechanical dissociation isolates neonatal and adult astrocytes better than enzymes. Papain + dispase II alows cell cytometry quantification of glial activation by LPS.
The technical difficulty to isolate microglia, astrocytes and infiltrating immune cells from mouse brain is nowadays a limiting factor in the study of neuroinflammation. Brain isolation requirements are cell-type and animal-age dependent, but current brain dissociation procedures are poorly standardized. This lack of comprehensive studies hampers the selection of optimized methodologies. Thus, we present here a comparative analysis of dissociation methods and Percoll-based separation to identify the most efficient procedure for the combined isolation of healthy microglia, astrocytes and infiltrated leukocytes; distinguishing neonatal and adult mouse brain. Gentle mechanical dissociation and DNase I incubation was supplemented with papain or collagenase II. Dispase II digestion was also used alone or in combination. In addition, cell separation efficiency of 30 % and 30–70 % Percoll gradients was compared. In these experiments, cell yield and integrity of freshly dissociated cells was measured by flow cytometry. We found that papain digestion in combination with dispase II followed by 30 % Percoll separation is the most balanced method to obtain a mixture of microglia, astrocytes and infiltrated immune cells; while addition of dispase II was not an advantage for neonatal brain. These dissociation conditions allowed flow cytometry detection of a slight glial activation triggered by sublethal LPS injection. In conclusion, the enzymes and Percoll density gradients tested here affected differently resting microglia, activated microglia/macrophages, astrocytes and infiltrated lymphocytes. Also, newborn and adult brain showed contrasting reactions to digestion. Our study highlights the strength of flow cytometry for the simultaneous analysis of neuroimmune cell populations once extraction is optimized.
Collapse
Key Words
- ANOVA, one-way analysis of variance
- Astrocytes
- CNS, Central Nervous System
- CaCl2, calcium chloride
- EBSS, Earle's Balanced Salt Solution
- EDTA, ethylenediaminetetraacetic acid
- FACS, Fluorescence-activated cell sorter
- FSC, forward-scattered light
- Flow cytometry
- Glia reactivity
- HBSS, Hank's Balanced Salt Solution
- LD, lethal dose
- LPS, lipopolysaccharide
- Lymphocytes
- MgCl2, magnesium chloride
- MgSO4, magnesium sulfate
- Microglia
- Neuroimmunity
- PBS, phosphate-buffered saline
- RT, room temperature
- SIP, stock solution of isotonic Percoll
- SSC, side-scattered light
- i.p, intraperitoneal injection
Collapse
Affiliation(s)
- Belén Calvo
- Neuroglia Laboratory, Research Institute for Neurological Disorders (IDINE), Medical School, University of Castilla-La Mancha (UCLM), Albacete, Spain
| | - Felipe Rubio
- Neuroglia Laboratory, Research Institute for Neurological Disorders (IDINE), Medical School, University of Castilla-La Mancha (UCLM), Albacete, Spain
| | - Miriam Fernández
- Neuroglia Laboratory, Research Institute for Neurological Disorders (IDINE), Medical School, University of Castilla-La Mancha (UCLM), Albacete, Spain
| | - Pedro Tranque
- Neuroglia Laboratory, Research Institute for Neurological Disorders (IDINE), Medical School, University of Castilla-La Mancha (UCLM), Albacete, Spain
| |
Collapse
|
32
|
Pedersbæk D, Jønsson K, Madsen DV, Weller S, Bohn AB, Andresen TL, Simonsen JB. A quantitativeex vivostudy of the interactions between reconstituted high-density lipoproteins and human leukocytes. RSC Adv 2020; 10:3884-3894. [PMID: 35492676 PMCID: PMC9048990 DOI: 10.1039/c9ra08203d] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/11/2020] [Indexed: 12/12/2022] Open
Abstract
Knowledge of the interactions between nanoparticles and immune cells is required for optimal design of nanoparticle-based drug delivery systems, either when aiming to avoid phagocytic clearance of the nanoparticles or promote an immune response by delivering therapeutic agents to specific immune cells. Several studies have suggested that reconstituted high-density lipoproteins (rHDL) are attractive drug delivery vehicles. However, detailed studies of rHDL interactions with circulating leukocytes are limited. Here, we evaluated the association of discoidal rHDL with leukocytes in human whole blood (HWB) using quantitative approaches. We found that while the rHDL of various lipid compositions associated preferentially with monocytes, the degree of association depended on the lipid composition. However, consistent with the long circulation half-life of rHDL, we show that only a minor fraction of the rHDL associated with the leukocytes. Furthermore, we used three-dimensional fluorescence microscopy and imaging flow cytometry to evaluate the possible internalization of rHDL cargo into the cells, and we show increased internalization of rHDL cargo in monocytes relative to granulocytes. The preferential rHDL association with monocytes and the internalization of rHDL cargo could possibly be mediated by the scavenger receptor class B type 1 (SR-BI), which we show is expressed to a higher extent on monocytes than on the other major leukocyte populations. Our work implies that drug-loaded rHDL can deliver its cargo to monocytes in circulation, which could lead to some off-target effects when using rHDL for systemic drug delivery, or it could pave the way for novel immunotherapeutic treatments aiming to target the monocytes. We used novel quantitative methods to study the interactions between reconstituted high-density lipoproteins (rHDL) and human leukocytes – showing that rHDL cargo are preferentially taken up by monocytes.![]()
Collapse
Affiliation(s)
- Dennis Pedersbæk
- Department of Health Technology
- Technical University of Denmark
- 2800 Kongens Lyngby
- Denmark
| | - Katrine Jønsson
- Department of Health Technology
- Technical University of Denmark
- 2800 Kongens Lyngby
- Denmark
| | - Ditte V. Madsen
- Department of Health Technology
- Technical University of Denmark
- 2800 Kongens Lyngby
- Denmark
| | - Sven Weller
- Department of Health Technology
- Technical University of Denmark
- 2800 Kongens Lyngby
- Denmark
| | - Anja B. Bohn
- Department of Biomedicine
- Aarhus University
- 8000 Aarhus
- Denmark
| | - Thomas L. Andresen
- Department of Health Technology
- Technical University of Denmark
- 2800 Kongens Lyngby
- Denmark
| | - Jens B. Simonsen
- Department of Health Technology
- Technical University of Denmark
- 2800 Kongens Lyngby
- Denmark
| |
Collapse
|
33
|
Benevides L, Saltarelli VM, Pioto F, Sacramento LA, Dias MS, Rodríguez GR, Viola JPB, Carregaro V, Silva JS. NFAT1 Regulates Ly6C hi Monocyte Recruitment to the CNS and Plays an Essential Role in Resistance to Toxoplasma gondii Infection. Front Immunol 2019; 10:2105. [PMID: 31555297 PMCID: PMC6742953 DOI: 10.3389/fimmu.2019.02105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 08/21/2019] [Indexed: 12/24/2022] Open
Abstract
Monocytes play key roles in the maintenance of homeostasis and in the control of the infection. Monocytes are recruited from the bone marrow to inflammatory sites and are essential for antimicrobial activity to limit tissue damage and promote adaptive T cell responses. Here, we investigated the role of Nuclear Factor of Activated T cells 1 (NFAT1) in the regulation of Ly6Chi inflammatory monocyte recruitment to the CNS upon T. gondii infection. We show that NFAT-1-deficient monocytes are unable to migrate to the CNS of T. gondii-infected mice. Moreover, NFAT1−/− mice are highly susceptible to chronic T. gondii infection due to a failure to control parasite replication in the CNS. The inhibition of Ly6Chi inflammatory monocyte recruitment to the CNS severely blocked CXCL10 production and consequently the migration of IFN-γ-producing CD4+ T cells. Moreover, the transfer of Ly6Chi monocytes to infected NFAT1−/− mice favored CD4+ T cell migration to the CNS and resulted in the inhibition of parasite replication and host defense. Together, these results demonstrated for the first time the contribution of NFAT1 to the regulation of Ly6Chi monocyte recruitment to the CNS and to resistance during chronic T. gondii infection.
Collapse
Affiliation(s)
- Luciana Benevides
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Fiocruz-Bi-Institutional Translational Medicine Plataform, Ribeirão Preto, Brazil
| | - Verônica M Saltarelli
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Franciele Pioto
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Fiocruz-Bi-Institutional Translational Medicine Plataform, Ribeirão Preto, Brazil
| | - Laís A Sacramento
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Murilo S Dias
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Gretel R Rodríguez
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - João P B Viola
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Vanessa Carregaro
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - João S Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Fiocruz-Bi-Institutional Translational Medicine Plataform, Ribeirão Preto, Brazil
| |
Collapse
|
34
|
Hirako IC, Assis PA, Galvão-Filho B, Luster AD, Antonelli LR, Gazzinelli RT. Monocyte-derived dendritic cells in malaria. Curr Opin Microbiol 2019; 52:139-150. [PMID: 31542508 DOI: 10.1016/j.mib.2019.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 08/03/2019] [Accepted: 08/17/2019] [Indexed: 12/16/2022]
Abstract
The pathogenesis of malaria is a multifactorial syndrome associated with a deleterious inflammatory response that is responsible for many of the clinical manifestations. While dendritic cells (DCs) play a critical role in initiating acquired immunity and host resistance to infection, they also play a pathogenic role in inflammatory diseases. In our recent studies, we found in different rodent malaria models that the monocyte-derived DCs (MO-DCs) become, transiently, a main DC population in spleens and inflamed non-lymphoid organs. These studies suggest that acute infection with Plasmodium berghei promotes the differentiation of splenic monocytes into inflammatory monocytes (iMOs) and thereafter into MO-DCs that play a pathogenic role by promoting inflammation and tissue damage. The recruitment of MO-DCs to the lungs and brain are dependent on expression of CCR4 and CCR5, respectively, and expression of respective chemokine ligands in each organ. Once they reach the target organ the MO-DCs produce the CXCR3 ligands (CXCL9 and CXCL10), recruit CD8+ T cells, and produce toxic metabolites that play an important role in the development of experimental cerebral malaria (ECM) and acute respiratory distress syndrome (ARDS).
Collapse
Affiliation(s)
- Isabella C Hirako
- Fundação Oswaldo Cruz - Minas, 30190-002 Belo Horizonte, MG, Brazil; University of Massachusetts Medical School, 01605 Worcester, MA, United States
| | - Patrícia A Assis
- University of Massachusetts Medical School, 01605 Worcester, MA, United States
| | | | - Andrew D Luster
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Lis Rv Antonelli
- Fundação Oswaldo Cruz - Minas, 30190-002 Belo Horizonte, MG, Brazil
| | - Ricardo T Gazzinelli
- Fundação Oswaldo Cruz - Minas, 30190-002 Belo Horizonte, MG, Brazil; University of Massachusetts Medical School, 01605 Worcester, MA, United States; Plataforma de Medicina Translacional, Fundação Oswaldo Cruz, 14049-900, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
35
|
Limitations of neutrophil depletion by anti-Ly6G antibodies in two heterogenic immunological models. Immunol Lett 2019; 212:30-36. [DOI: 10.1016/j.imlet.2019.06.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/24/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023]
|
36
|
Lakhdari O, Yamamura A, Hernandez GE, Anderson KK, Lund SJ, Oppong-Nonterah GO, Hoffman HM, Prince LS. Differential Immune Activation in Fetal Macrophage Populations. Sci Rep 2019; 9:7677. [PMID: 31118442 PMCID: PMC6531440 DOI: 10.1038/s41598-019-44181-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/02/2019] [Indexed: 02/06/2023] Open
Abstract
Distinct macrophage subsets populate the developing embryo and fetus in distinct waves. However little is known about the functional differences between in utero macrophage populations or how they might contribute to fetal and neonatal immunity. Here we tested the innate immune response of mouse macrophages derived from the embryonic yolk sac and from fetal liver. When isolated from liver or lung, CD11bHI fetal liver derived macrophages responded to the TLR4 agonist LPS by expressing and releasing inflammatory cytokines. However F4/80HI macrophages from the yolk sac did not respond to LPS treatment. While differences in TLR4 expression did not appear to explain these data, F4/80HI macrophages had much lower NLRP3 inflammasome expression compared to CD11bHI macrophages. Gene expression profiling also demonstrated LPS-induced expression of inflammatory genes in CD11bHI macrophages, but not in F4/80HI cells. Genes expressed in LPS-treated CD11bHI macrophages were more likely to contain predicted NF-κB binding sites in their promoter regions. Our data show that CD11bHI macrophages derived from fetal liver are the major pro-inflammatory cells in the developing fetus. These findings could have important implications in better understanding the fetal inflammatory response and the unique features of neonatal immunity.
Collapse
Affiliation(s)
- Omar Lakhdari
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, Rady Children's Hospital, San Diego, San Diego, CA, USA
| | - Asami Yamamura
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, Rady Children's Hospital, San Diego, San Diego, CA, USA
| | - Gilberto E Hernandez
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, Rady Children's Hospital, San Diego, San Diego, CA, USA
| | - Kathryn K Anderson
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, Rady Children's Hospital, San Diego, San Diego, CA, USA
| | - Sean J Lund
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, Rady Children's Hospital, San Diego, San Diego, CA, USA
| | - Gertrude O Oppong-Nonterah
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, Rady Children's Hospital, San Diego, San Diego, CA, USA
| | - Hal M Hoffman
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, Rady Children's Hospital, San Diego, San Diego, CA, USA
| | - Lawrence S Prince
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, Rady Children's Hospital, San Diego, San Diego, CA, USA.
| |
Collapse
|
37
|
Hortle E, Starrs L, Brown FC, Jane SM, Curtis DJ, McMorran BJ, Foote SJ, Burgio G. KCC1 Activation protects Mice from the Development of Experimental Cerebral Malaria. Sci Rep 2019; 9:6356. [PMID: 31015511 PMCID: PMC6478876 DOI: 10.1038/s41598-019-42782-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 04/08/2019] [Indexed: 11/15/2022] Open
Abstract
Plasmodium falciparum malaria causes half a million deaths per year, with up to 9% of this mortality caused by cerebral malaria (CM). One of the major processes contributing to the development of CM is an excess of host inflammatory cytokines. Recently K+ signaling has emerged as an important mediator of the inflammatory response to infection; we therefore investigated whether mice carrying an ENU induced activation of the electroneutral K+ channel KCC1 had an altered response to Plasmodium berghei. Here we show that Kcc1M935K/M935K mice are protected from the development of experimental cerebral malaria, and that this protection is associated with an increased CD4+ and TNFa response. This is the first description of a K+ channel affecting the development of experimental cerebral malaria.
Collapse
Affiliation(s)
- Elinor Hortle
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Australian Capital Territory, Australia
| | - Lora Starrs
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Australian Capital Territory, Australia
| | - Fiona C Brown
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Australia
| | - Stephen M Jane
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Australia.,The Alfred Hospital, Melbourne, Australia.,Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - David J Curtis
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Australia.,The Alfred Hospital, Melbourne, Australia
| | - Brendan J McMorran
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Australian Capital Territory, Australia
| | - Simon J Foote
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Australian Capital Territory, Australia
| | - Gaetan Burgio
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Australian Capital Territory, Australia.
| |
Collapse
|
38
|
Sierro F, Grau GER. The Ins and Outs of Cerebral Malaria Pathogenesis: Immunopathology, Extracellular Vesicles, Immunometabolism, and Trained Immunity. Front Immunol 2019; 10:830. [PMID: 31057552 PMCID: PMC6478768 DOI: 10.3389/fimmu.2019.00830] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 03/28/2019] [Indexed: 12/16/2022] Open
Abstract
Complications from malaria parasite infections still cost the lives of close to half a million people every year. The most severe is cerebral malaria (CM). Employing murine models of CM, autopsy results, in vitro experiments, neuroimaging and microscopic techniques, decades of research activity have investigated the development of CM immunopathology in the hope of identifying steps that could be therapeutically targeted. Yet important questions remain. This review summarizes recent findings, primarily mechanistic insights on the essential cellular and molecular players involved gained within the murine experimental cerebral malaria model. It also highlights recent developments in (a) cell-cell communication events mediated through extracellular vesicles (EVs), (b) mounting evidence for innate immune memory, leading to “trained“ increased or tolerised responses, and (c) modulation of immune cell function through metabolism, that could shed light on why some patients develop this life-threatening condition whilst many do not.
Collapse
Affiliation(s)
- Frederic Sierro
- Vascular Immunology Unit, Department of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia.,Human Health, Nuclear Science, Technology, and Landmark Infrastructure, Australian Nuclear Science and Technology Organisation, Sydney, NSW, Australia
| | - Georges E R Grau
- Vascular Immunology Unit, Department of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
39
|
Palomo J, Quesniaux VFJ, Togbe D, Reverchon F, Ryffel B. Unravelling the roles of innate lymphoid cells in cerebral malaria pathogenesis. Parasite Immunol 2019; 40. [PMID: 29117626 DOI: 10.1111/pim.12502] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 10/31/2017] [Indexed: 12/17/2022]
Abstract
Cerebral malaria (CM) is one complication of Plasmodium parasite infection that can lead to strong inflammatory immune responses in the central nervous system (CNS), accompanied by lung inflammation and anaemia. Here, we focus on the role of the innate immune response in experimental cerebral malaria (ECM) caused by blood-stage murine Plasmodium berghei ANKA infection. While T cells are important for ECM pathogenesis, the role of innate lymphoid cells (ILCs) is only emerging. The role of ILCs and non-lymphoid cells, such as neutrophils and platelets, contributing to the host immune response and leading to ECM and human cerebral malaria (HCM) is reviewed.
Collapse
Affiliation(s)
- J Palomo
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS, University of Orleans, Orleans-Cedex2, France.,Division of Rheumatology, Departments of Internal Medicine Specialties and of Pathology-Immunology, University of Geneva School of Medicine, Geneva, Switzerland
| | - V F J Quesniaux
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS, University of Orleans, Orleans-Cedex2, France
| | - D Togbe
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS, University of Orleans, Orleans-Cedex2, France.,Artimmune SAS, Orléans, France
| | - F Reverchon
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS, University of Orleans, Orleans-Cedex2, France
| | - B Ryffel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS, University of Orleans, Orleans-Cedex2, France.,IDM, Medical School, University of Cape Town, Cape Town, Republic of South Africa
| |
Collapse
|
40
|
Richey T, Foster JS, Williams AD, Williams AB, Stroh A, Macy S, Wooliver C, Heidel RE, Varanasi SK, Ergen EN, Trent DJ, Kania SA, Kennel SJ, Martin EB, Wall JS. Macrophage-Mediated Phagocytosis and Dissolution of Amyloid-Like Fibrils in Mice, Monitored by Optical Imaging. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:989-998. [PMID: 30735627 DOI: 10.1016/j.ajpath.2019.01.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/14/2019] [Accepted: 01/17/2019] [Indexed: 01/27/2023]
Abstract
Light chain-associated amyloidosis is characterized by the extracellular deposition of amyloid fibrils in abdominothoracic organs, skin, soft tissue, and peripheral nerves. Phagocytic cells of the innate immune system appear to be ineffective at clearing the material; however, human light chain amyloid extract, injected subcutaneously into mice, is rapidly cleared in a process that requires neutrophil activity. To better elucidate the phagocytosis of light chain fibrils, a potential method of cell-mediated dissolution, amyloid-like fibrils were labeled with the pH-sensitive dye pHrodo red and a near infrared fluorophore. After injecting this material subcutaneously in mice, optical imaging was used to quantitatively monitor phagocytosis and dissolution of fibrils concurrently. Histologic evaluation of the residual fibril masses revealed the presence of CD68+, F4/80+, ionized calcium binding adaptor molecule 1- macrophages containing Congo red-stained fibrils as well as neutrophil-associated proteins with no evidence of intact neutrophils. These data suggest an early infiltration of neutrophils, followed by extensive phagocytosis of the light chain fibrils by macrophages, leading to dissolution of the mass. Optical imaging of this novel murine model, coupled with histologic evaluation, can be used to study the cellular mechanisms underlying dissolution of synthetic amyloid-like fibrils and human amyloid extracts. In addition, it may serve as a test bed to evaluate investigational opsonizing agents that might serve as therapeutic agents for light chain-associated amyloidosis.
Collapse
Affiliation(s)
- Tina Richey
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee
| | - James S Foster
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee
| | - Angela D Williams
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee
| | | | - Alexa Stroh
- Department of Biochemistry, Cellular and Molecular Biology, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee
| | - Sallie Macy
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee
| | - Craig Wooliver
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee
| | - R Eric Heidel
- Department of Surgery, University of Tennessee Medical Center, Knoxville, Tennessee
| | - Siva K Varanasi
- Department of Biochemistry, Cellular and Molecular Biology, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee
| | - Elizabeth N Ergen
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee
| | - Dianne J Trent
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee
| | - Stephen A Kania
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee
| | - Stephen J Kennel
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee
| | - Emily B Martin
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee
| | - Jonathan S Wall
- Department of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee.
| |
Collapse
|
41
|
Niewold P, Cohen A, van Vreden C, Getts DR, Grau GE, King NJC. Experimental severe malaria is resolved by targeting newly-identified monocyte subsets using immune-modifying particles combined with artesunate. Commun Biol 2018; 1:227. [PMID: 30564748 PMCID: PMC6292940 DOI: 10.1038/s42003-018-0216-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/31/2018] [Indexed: 12/29/2022] Open
Abstract
Current treatment of severe malaria and associated cerebral malaria (CM) and respiratory distress syndromes are directed primarily at the parasite. Targeting the parasite has only partial efficacy in advanced infection, as neurological damage and respiratory distress are due to accumulation of host blood cells in the brain microvasculature and lung interstitium. Here, computational analysis identifies Ly6Clo monocytes as a major component of the immune infiltrate in both organs in a preclinical mouse model. Specifically targeting Ly6Clo monocyte precursors, identified by adoptive transfer, with immune-modifying particles (IMP) prevents experimental CM (ECM) in 50% of Plasmodium berghei ANKA-infected mice in early treatment protocols. Furthermore, treatment at onset of clinical ECM with 2 doses of a novel combination of IMP and anti-malarial drug artesunate results in 88% survival. This combination confers protection against ECM and mortality in late stage severe experimental malaria and provides a viable advance on current treatment regimens.
Collapse
Affiliation(s)
- Paula Niewold
- 1Viral Immunopathology, Discipline of Pathology and Bosch Institute, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050 Australia
| | - Amy Cohen
- 2Vascular Immunology Unit, Discipline of Pathology and Bosch Institute, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050 Australia
| | - Caryn van Vreden
- 3Sydney Cytometry, The University of Sydney and The Centenary Institute, Camperdown, NSW 2050 Australia
| | - Daniel R Getts
- 4Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611 USA.,TcR2, Therapeutics, 100 Binney Street, Suite 710, Cambridge, MA 02142 USA
| | - Georges E Grau
- 2Vascular Immunology Unit, Discipline of Pathology and Bosch Institute, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050 Australia
| | - Nicholas J C King
- 1Viral Immunopathology, Discipline of Pathology and Bosch Institute, School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050 Australia.,3Sydney Cytometry, The University of Sydney and The Centenary Institute, Camperdown, NSW 2050 Australia
| |
Collapse
|
42
|
Peñaloza HF, Alvarez D, Muñoz-Durango N, Schultz BM, González PA, Kalergis AM, Bueno SM. The role of myeloid-derived suppressor cells in chronic infectious diseases and the current methodology available for their study. J Leukoc Biol 2018; 105:857-872. [PMID: 30480847 DOI: 10.1002/jlb.mr0618-233r] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 10/07/2018] [Accepted: 10/30/2018] [Indexed: 12/23/2022] Open
Abstract
An effective pathogen has the ability to evade the immune response. The strategies used to achieve this may be based on the direct action of virulence factors or on the induction of host factors. Myeloid-derived suppressor cells (MDSCs) are immune cells with an incredible ability to suppress the inflammatory response, which makes them excellent targets to be exploited by pathogenic bacteria, viruses, or parasites. In this review, we describe the origin and suppressive mechanisms of MDSCs, as well as their role in chronic bacterial, viral, and parasitic infections, where their expansion seems to be essential in the chronicity of the disease. We also analyze the disadvantages of current MDSC depletion strategies and the different in vitro generation methods, which can be useful tools for the deeper study of these cells in the context of microbial infections.
Collapse
Affiliation(s)
- Hernán F Peñaloza
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Diana Alvarez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Natalia Muñoz-Durango
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bárbara M Schultz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
43
|
Kreer C, Kuepper JM, Zehner M, Quast T, Kolanus W, Schumak B, Burgdorf S. N-glycosylation converts non-glycoproteins into mannose receptor ligands and reveals antigen-specific T cell responses in vivo. Oncotarget 2018; 8:6857-6872. [PMID: 28036287 PMCID: PMC5351675 DOI: 10.18632/oncotarget.14314] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 12/05/2016] [Indexed: 11/25/2022] Open
Abstract
N-glycosylation is generally accepted to enhance the immunogenicity of antigens because of two main reasons. First, the attachment of glycans enables recognition by endocytic receptors like the mannose receptor (MR) and hence increased uptake by dendritic cells (DCs). Second, foreign glycans are postulated to be immunostimulatory and their recognition could induce DC activation. However, a direct comparison between the immunogenicity of N-glycosylated vs. de-glycosylated proteins in vivo and a direct effect of N-glycosylated antigens on the intrinsic capacity of DCs to activate T cells have not been assessed so far.To analyze whether enforced N-glycosylation is a suited strategy to enhance the immunogenicity of non-glycosylated antigens for vaccination studies, we targeted non-glycoproteins towards the MR by introduction of artificial N-glycosylation using the methylotrophic yeast Komagataella phaffii (previously termed Pichia pastoris). We could demonstrate that the introduction of a single N-X-S/T motif was sufficient for efficient MR-binding and internalization. However, addition of N-glycosylated proteins neither influenced DC maturation nor their general capacity to activate T cells, pointing out that enforced N-glycosylation does not increase the immunogenicity of the antigen per se. Additionally, increased antigen-specific cytotoxic T cell responses in vivo after injection of N-glycosylated compared to de-glycosylated proteins were observed but this effect strongly depended on the epitope tested. A beneficial effect of N-glycosylation on antibody production could not be detected, which might be due to MR-cross-linking on DCs and to concomitant differences in IL-6 production by CD4+ T cells.These observations point out that the effect of N-glycosylation on antigen immunogenicity can vary between different antigens and therefore might have important implications for the development of vaccines using K. phaffii.
Collapse
Affiliation(s)
- Christoph Kreer
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany
| | - Janina M Kuepper
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53105 Bonn, Germany
| | - Matthias Zehner
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany
| | - Thomas Quast
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany
| | - Waldemar Kolanus
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany
| | - Beatrix Schumak
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, 53105 Bonn, Germany
| | - Sven Burgdorf
- Life and Medical Sciences (LIMES) Institute, University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
44
|
Riggle BA, Miller LH, Pierce SK. Do we know enough to find an adjunctive therapy for cerebral malaria in African children? F1000Res 2017; 6:2039. [PMID: 29250318 PMCID: PMC5701444 DOI: 10.12688/f1000research.12401.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/23/2017] [Indexed: 01/27/2023] Open
Abstract
Cerebral malaria is the deadliest complication of malaria, a febrile infectious disease caused by Plasmodium parasite. Any of the five human Plasmodium species can cause disease, but, for unknown reasons, in approximately 2 million cases each year P. falciparum progresses to severe disease, ultimately resulting in half a million deaths. The majority of these deaths are in children under the age of five. Currently, there is no way to predict which child will progress to severe disease and there are no adjunctive therapies to halt the symptoms after onset. Herein, we discuss what is known about the disease mechanism of one form of severe malaria, cerebral malaria, and how we might exploit this understanding to rescue children in the throes of cerebral disease.
Collapse
Affiliation(s)
- Brittany A Riggle
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, USA
| | - Louis H Miller
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, USA
| | - Susan K Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, USA
| |
Collapse
|
45
|
Effects of Acyclovir and IVIG on Behavioral Outcomes after HSV1 CNS Infection. Behav Neurol 2017; 2017:5238402. [PMID: 29358844 PMCID: PMC5735307 DOI: 10.1155/2017/5238402] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/06/2017] [Accepted: 09/16/2017] [Indexed: 12/27/2022] Open
Abstract
Herpes simplex virus 1 (HSV) encephalitis (HSE) has serious neurological complications, involving behavioral and cognitive impairments that cause significant morbidity and a reduced quality of life. We showed that HSE results from dysregulated central nervous system (CNS) inflammatory responses. We hypothesized that CNS inflammation is casually involved in behavioral abnormalities after HSE and that treatment with ACV and pooled human immunoglobulin (IVIG), an immunomodulatory drug, would improve outcomes compared to mice treated with phosphate buffered saline (PBS) or ACV alone. Anxiety levels were high in HSV-infected PBS and ACV-treated mice compared to mice treated with ACV + IVIG, consistent with reports implicating inflammation in anxiety induced by lipopolysaccharide (LPS) or stress. Female, but not male, PBS-treated mice were cognitively impaired, and unexpectedly, ACV was protective, while the inclusion of IVIG surprisingly antagonized ACV's beneficial effects. Distinct serum proteomic profiles were observed for male and female mice, and the antagonistic effects of ACV and IVIG on behavior were paralleled by similar changes in the serum proteome of ACV- and ACV + IVIG-treated mice. We conclude that inflammation and other factors mediate HSV-induced behavioral impairments and that the effects of ACV and IVIG on behavior involve novel mechanisms.
Collapse
|
46
|
Inflammatory monocytes regulate Th1 oriented immunity to CpG adjuvanted protein vaccines through production of IL-12. Sci Rep 2017; 7:5986. [PMID: 28729715 PMCID: PMC5519561 DOI: 10.1038/s41598-017-06236-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 06/12/2017] [Indexed: 01/26/2023] Open
Abstract
Due to their capacity to skew T cell responses towards Th1 oriented immunity, oligonucleotides containing unmethylated CpG motifs (CpG) have emerged as interesting adjuvants for vaccination. Whereas the signalling pathways in response to CpG mediated TLR9 activation have been extensively documented at the level of the individual cell, little is however known on the precise identity of the innate immune cells that govern T cell priming and polarisation to CpG adjuvanted protein antigens in vivo. In this study, we demonstrate that optimal induction of Th1 oriented immunity to CpG adjuvanted protein vaccines requires the coordinated actions of conventional DCs and of monocytes. Whilst conventional DCs were required for antigen presentation and initial T cell priming, monocytes constitute the main source of the Th1 polarising cytokine IL-12.
Collapse
|
47
|
Wu X, Dayanand KK, Thylur RP, Norbury CC, Gowda DC. Small molecule-based inhibition of MEK1/2 proteins dampens inflammatory responses to malaria, reduces parasite load, and mitigates pathogenic outcomes. J Biol Chem 2017; 292:13615-13634. [PMID: 28679535 DOI: 10.1074/jbc.m116.770313] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 06/23/2017] [Indexed: 12/16/2022] Open
Abstract
Malaria infections cause several systemic and severe single- or multi-organ pathologies, killing hundreds of thousands of people annually. Considering the existing widespread resistance of malaria parasites to anti-parasitic drugs and their high propensity to develop drug resistance, alternative strategies are required to manage malaria infections. Because malaria is a host immune response-driven disease, one approach is based on gaining a detailed understanding of the molecular and cellular processes that modulate malaria-induced innate and adaptive immune responses. Here, using a mouse cerebral malaria model and small-molecule inhibitors, we demonstrate that inhibiting MEK1/2, the upstream kinases of ERK1/2 signaling, alters multifactorial components of the innate and adaptive immune responses, controls parasitemia, and blocks pathogenesis. Specifically, MEK1/2 inhibitor treatment up-regulated B1 cell expansion, IgM production, phagocytic receptor expression, and phagocytic activity, enhancing parasite clearance by macrophages and neutrophils. Further, the MEK1/2 inhibitor treatment down-regulated pathogenic pro-inflammatory and helper T cell 1 (Th1) responses and up-regulated beneficial anti-inflammatory cytokine responses and Th2 responses. These inhibitor effects resulted in reduced granzyme B expression by T cells, chemokine and intracellular cell adhesion molecule 1 (ICAM-1) expression in the brain, and chemokine receptor expression by both myeloid and T cells. These bimodal effects of the MEK1/2 inhibitor treatment on immune responses contributed to decreased parasite biomass, organ inflammation, and immune cell recruitment, preventing tissue damage and death. In summary, we have identified several previously unrecognized immune regulatory processes through which a MEK1/2 inhibitor approach controls malaria parasitemia and mitigates pathogenic effects on host organs.
Collapse
Affiliation(s)
- Xianzhu Wu
- From the Departments of Biochemistry and Molecular Biology and
| | | | - Ramesh P Thylur
- From the Departments of Biochemistry and Molecular Biology and
| | - Christopher C Norbury
- Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - D Channe Gowda
- From the Departments of Biochemistry and Molecular Biology and
| |
Collapse
|
48
|
Lee AJ, Chen B, Chew MV, Barra NG, Shenouda MM, Nham T, van Rooijen N, Jordana M, Mossman KL, Schreiber RD, Mack M, Ashkar AA. Inflammatory monocytes require type I interferon receptor signaling to activate NK cells via IL-18 during a mucosal viral infection. J Exp Med 2017; 214:1153-1167. [PMID: 28264883 PMCID: PMC5379971 DOI: 10.1084/jem.20160880] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 11/15/2016] [Accepted: 01/25/2017] [Indexed: 11/17/2022] Open
Abstract
Although type I interferon is critical for NK cell activation, the underlying mechanism is under debate and is unknown during a mucosal infection. Lee et al. have determined that type I interferon induces inflammatory monocytes to produce IL-18 to directly activate NK cells to combat viral infections. The requirement of type I interferon (IFN) for natural killer (NK) cell activation in response to viral infection is known, but the underlying mechanism remains unclear. Here, we demonstrate that type I IFN signaling in inflammatory monocytes, but not in dendritic cells (DCs) or NK cells, is essential for NK cell function in response to a mucosal herpes simplex virus type 2 (HSV-2) infection. Mice deficient in type I IFN signaling, Ifnar−/− and Irf9−/− mice, had significantly lower levels of inflammatory monocytes, were deficient in IL-18 production, and lacked NK cell–derived IFN-γ. Depletion of inflammatory monocytes, but not DCs or other myeloid cells, resulted in lower levels of IL-18 and a complete abrogation of NK cell function in HSV-2 infection. Moreover, this resulted in higher susceptibility to HSV-2 infection. Although Il18−/− mice had normal levels of inflammatory monocytes, their NK cells were unresponsive to HSV-2 challenge. This study highlights the importance of type I IFN signaling in inflammatory monocytes and the induction of the early innate antiviral response.
Collapse
Affiliation(s)
- Amanda J Lee
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Branson Chen
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Marianne V Chew
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Nicole G Barra
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Mira M Shenouda
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Tina Nham
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Nico van Rooijen
- Department of Molecular Cell Biology, Vrije University Medical Center, 1081 HV Amsterdam, Netherlands
| | - Manel Jordana
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Karen L Mossman
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | | | - Matthias Mack
- RCI Regensburg Center for Interventional Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Ali A Ashkar
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| |
Collapse
|
49
|
Chow KV, Carrington EM, Zhan Y, Lew AM, Sutherland RM. Monocyte-Derived Dendritic Cells Impair Early Graft Function Following Allogeneic Islet Transplantation. Cell Transplant 2017; 26:319-326. [PMID: 27743446 PMCID: PMC5657768 DOI: 10.3727/096368916x693482] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 12/08/2016] [Indexed: 12/18/2022] Open
Abstract
Islet transplantation can cure type 1 diabetes but is limited by lack of donor organs and early graft dysfunction, such that many patients require multiple transplants to achieve insulin independence. Monocyte-derived dendritic cells (moDCs) arise during inflammation and allograft encounters where they can promote various innate and adaptive immune responses. To determine whether moDCs impair early graft function following allogeneic islet transplantation, we transplanted MHC-mismatched BALB/c (H-2d) islets into diabetic C57BL/6-CCR2.DTR recipients (H-2b) treated with either saline (control) or diphtheria toxin (DT) to deplete moDCs. Graft function was assessed by blood glucose (BG) measurement. DT treatment resulted in specific depletion of graft site moDCs posttransplant. Despite equivalent pretransplant BG levels [27.0 ± 1.3 vs. 29.6 ± 1.1 mM, not significant (ns)], DT recipients achieved lower posttransplant BG levels and better rates of normoglycemia than control recipients (11.0 ± 1.9 vs. 19.1 ± 1.4 mM, p = 0.004) at 1 day posttransplant in diabetic recipients. When a suboptimal donor dose of 200 islets was transplanted, DT-induced moDC depletion resulted in normoglycemia in 78% compared to 25% of control recipients (p = 0.03). As well as amelioration of graft dysfunction in the immediate peritransplant period, prolonged DT administration (15 days posttransplant) resulted in improved graft survival (21 vs. 11 days, p = 0.005). moDCs impair early graft function post-allogeneic islet transplantation. moDC depletion may allow for improved early graft function, permit transplantation with lower islet masses, and enhance graft survival.
Collapse
Affiliation(s)
- Kevin V. Chow
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
- Department of Nephrology, The Royal Melbourne Hospital, Parkville, Australia
| | - Emma M. Carrington
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Yifan Zhan
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Andrew M. Lew
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
- Department of Microbiology and Immunology, University of Melbourne, Parkville, Australia
| | - Robyn M. Sutherland
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| |
Collapse
|
50
|
Galbas T, Raymond M, Sabourin A, Bourgeois-Daigneault MC, Guimont-Desrochers F, Yun TJ, Cailhier JF, Ishido S, Lesage S, Cheong C, Thibodeau J. MARCH1 E3 Ubiquitin Ligase Dampens the Innate Inflammatory Response by Modulating Monocyte Functions in Mice. THE JOURNAL OF IMMUNOLOGY 2016; 198:852-861. [PMID: 27940660 DOI: 10.4049/jimmunol.1601168] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 11/09/2016] [Indexed: 12/15/2022]
Abstract
Ubiquitination was recently identified as a central process in the pathogenesis and development of numerous inflammatory diseases, such as obesity, atherosclerosis, and asthma. Treatment with proteasomal inhibitors led to severe side effects because ubiquitination is heavily involved in a plethora of cellular functions. Thus, new players regulating ubiquitination processes must be identified to improve therapies for inflammatory diseases. In addition to their role in adaptive immunity, endosomal MHC class II (MHCII) molecules were shown to modulate innate immune responses by fine tuning the TLR4 signaling pathway. However, the role of MHCII ubiquitination by membrane associated ring-CH-type finger 1 (MARCH1) E3 ubiquitin ligase in this process remains to be assessed. In this article, we demonstrate that MARCH1 is a key inhibitor of innate inflammation in response to bacterial endotoxins. The higher mortality of March1-/- mice challenged with a lethal dose of LPS was associated with significantly stronger systemic production of proinflammatory cytokines and splenic NK cell activation; however, we did not find evidence that MARCH1 modulates LPS or IL-10 signaling pathways. Instead, the mechanism by which MARCH1 protects against endotoxic shock rests on its capacity to promote the transition of monocytes from Ly6CHi to Ly6C+/- Moreover, in competitive bone marrow chimeras, March1-/- monocytes and polymorphonuclear neutrophils outcompeted wild-type cells with regard to bone marrow egress and homing to peripheral organs. We conclude that MARCH1 exerts MHCII-independent effects that regulate the innate arm of immunity. Thus, MARCH1 might represent a potential new target for emerging therapies based on ubiquitination reactions in inflammatory diseases.
Collapse
Affiliation(s)
- Tristan Galbas
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montreal, Montreal, Quebec H3T 1J4, Canada.,Laboratoire d'Immunologie Moléculaire, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Maxime Raymond
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montreal, Montreal, Quebec H3T 1J4, Canada.,Laboratoire d'Immunologie Moléculaire, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Antoine Sabourin
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montreal, Montreal, Quebec H3T 1J4, Canada.,Laboratoire d'Immunologie Moléculaire, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Marie-Claude Bourgeois-Daigneault
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montreal, Montreal, Quebec H3T 1J4, Canada.,Laboratoire d'Immunologie Moléculaire, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Fanny Guimont-Desrochers
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montreal, Montreal, Quebec H3T 1J4, Canada.,Immunology-Oncology Section, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada
| | - Tae Jin Yun
- Laboratoire de Physiologie Cellulaire et Immunologie, Institut de Recherches Cliniques de Montréal, Montreal, Quebec H2W 1R7, Canada
| | - Jean-François Cailhier
- Centre de Recherche, Centre Hospitalier de l'Université de Montréal, Montreal, Quebec H2X 0A9, Canada; and
| | - Satoshi Ishido
- Department of Microbiology, Hyogo College of Medicine 1-1, Mukogawa-cho, Nishinomiya 663-8501, Japan
| | - Sylvie Lesage
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montreal, Montreal, Quebec H3T 1J4, Canada.,Immunology-Oncology Section, Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada
| | - Cheolho Cheong
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montreal, Montreal, Quebec H3T 1J4, Canada.,Laboratoire de Physiologie Cellulaire et Immunologie, Institut de Recherches Cliniques de Montréal, Montreal, Quebec H2W 1R7, Canada
| | - Jacques Thibodeau
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montreal, Montreal, Quebec H3T 1J4, Canada; .,Laboratoire d'Immunologie Moléculaire, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| |
Collapse
|