1
|
Hsu AY, Huang Q, Pi X, Fu J, Raghunathan K, Ghimire L, Balasubramanian A, Xie X, Yu H, Loison F, Haridas V, Zha J, Liu F, Park SY, Bagale K, Ren Q, Fan Y, Zheng Y, Cancelas JA, Chai L, Stowell SR, Chen K, Xu R, Wang X, Xu Y, Zhang L, Cheng T, Ma F, Thiagarajah JR, Wu H, Feng S, Luo HR. Neutrophil-derived vesicles control complement activation to facilitate inflammation resolution. Cell 2025; 188:1623-1641.e26. [PMID: 39938514 PMCID: PMC11934499 DOI: 10.1016/j.cell.2025.01.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 08/23/2024] [Accepted: 01/13/2025] [Indexed: 02/14/2025]
Abstract
Although subsets with immunosuppressive properties exist, neutrophils are typically known for their pro-inflammatory role and pathogen clearance capabilities. Here, we reveal that neutrophils can paradoxically aid in resolving inflammation by actively producing anti-inflammatory extracellular vesicles. These large aging-neutrophil-derived vesicles (LAND-Vs) do not fit into classical vesicle categorizations due to their specific size, structure, or biogenesis pathway. They are protected from efferocytotic clearance by phagocytes due to surface "do not eat me" signals and accumulate in the resolution phase of inflammation. CD55 on LAND-Vs exerts a robust, sustained anti-inflammatory effect by inhibiting complement 3 convertase, thereby reducing neutrophil recruitment and tissue damage. CD55+ LAND-Vs originate in ordered lipid raft domains, where CD55 accumulates asymmetrically during neutrophil aging, and are subsequently formed through RhoA-dependent budding. Collectively, LAND-V emerges as a pivotal physiological immunomodulator and showcases functions that transcend the limited lifespan of neutrophils, offering a therapeutic target for inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Alan Y Hsu
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Qingxiang Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Xiong Pi
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 20115, USA
| | - Jianing Fu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 20115, USA
| | - Krishnan Raghunathan
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA 20115, USA
| | - Laxman Ghimire
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Arumugam Balasubramanian
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Xuemei Xie
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Hongbo Yu
- Department of Pathology and Laboratory Medicine, VA Boston Healthcare System, West Roxbury, Boston, MA 02132, USA
| | - Fabien Loison
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Viraga Haridas
- Flow and Imaging Cytometry Resources, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jiali Zha
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Fei Liu
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Shin-Young Park
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Kamal Bagale
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Qian Ren
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yuping Fan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yi Zheng
- Experimental Hematology and Cancer Biology Research, Cincinnati Children's Hospital Medical Center, Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Jose A Cancelas
- Experimental Hematology and Cancer Biology Research, Cincinnati Children's Hospital Medical Center, Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Li Chai
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Sean R Stowell
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Kanchao Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Rong Xu
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA
| | - Xiaoxue Wang
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yuanfu Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Lianghui Zhang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Fengxia Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Jay R Thiagarajah
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA 20115, USA; Congenital Enteropathy Program, Boston Children's Hospital, PediCODE Consortium, Harvard Digestive Disease Center, Boston, MA, USA
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 20115, USA
| | - Sizhou Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China.
| | - Hongbo R Luo
- Department of Pathology, PhD Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA; Department of Pathology, Mass General Brigham, Boston, MA 02115, USA.
| |
Collapse
|
2
|
An H, Huang Y, Zhao Z, Li K, Meng J, Huang X, Tian X, Zhou H, Wu J, Dai Q, Zhang JR. Splenic red pulp macrophages eliminate the liver-resistant Streptococcus pneumoniae from the blood circulation of mice. SCIENCE ADVANCES 2025; 11:eadq6399. [PMID: 40073120 PMCID: PMC11900858 DOI: 10.1126/sciadv.adq6399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025]
Abstract
Invasive infections by encapsulated bacteria are the major cause of human morbidity and mortality. The liver resident macrophages, Kupffer cells, form the hepatic firewall to clear many encapsulated bacteria in the blood circulation but fail to control certain high-virulence capsule types. Here we report that the spleen is the backup immune organ to clear the liver-resistant serotypes of Streptococcus pneumoniae (pneumococcus), a leading human pathogen. Asplenic mice failed to control the growth of the liver-resistant pneumococci in the blood circulation. Immunologic and genetic analyses identified splenic red pulp (RP) macrophages as the major phagocytes for bacterial clearance. Furthermore, the plasma natural antibodies against the cell wall phosphocholine and the complement system were necessary for RP macrophage-mediated immunity. These findings have provided a conceptual framework for the innate defense against blood bacterial infections, a mechanistic explanation for the hyper-susceptibility of asplenic individuals to S. pneumoniae, and a proof of concept for developing vaccines and therapeutic antibodies against encapsulated pathogens.
Collapse
Affiliation(s)
- Haoran An
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China
- Department of Microbiology and Infectious Disease Center, Peking University Health Science Center, Beijing 100191, China
- Center for Infectious Biology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Yijia Huang
- Department of Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhifeng Zhao
- Department of Automation, Tsinghua University, Beijing 100084, China
| | - Kunpeng Li
- Center for Infectious Biology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Jingjing Meng
- Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China
| | - Xueting Huang
- Center for Infectious Biology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Xianbin Tian
- Center for Infectious Biology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Hongyu Zhou
- Department of Automation, Tsinghua University, Beijing 100084, China
| | - Jiamin Wu
- Department of Automation, Tsinghua University, Beijing 100084, China
| | - Qionghai Dai
- Department of Automation, Tsinghua University, Beijing 100084, China
| | - Jing-Ren Zhang
- Center for Infectious Biology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
3
|
Artola-Borán M, Kirsche L, Fallegger A, Leary P, Tanriover M, Goodwin T, Geiger G, Hapfelmeier S, Yousefi S, Simon HU, Arnold IC, Müller A. IgA facilitates the persistence of the mucosal pathogen Helicobacter pylori. Mucosal Immunol 2025; 18:232-247. [PMID: 39581230 DOI: 10.1016/j.mucimm.2024.11.006] [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: 07/01/2024] [Revised: 10/29/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024]
Abstract
IgA antibodies have an important role in clearing mucosal pathogens. In this study, we have examined the contribution of IgA to the immune control of the gastrointestinal bacterial pathogens Helicobacter pylori and Citrobacter rodentium. Both bacteria trigger a strong local IgA response that results in bacterial IgA coating in mice and in gastritis patients. Class switching to IgA depends on Peyer's patches, T-cells, eosinophils, and eosinophil-derived TGF-β in both models. In the case of H. pylori, IgA secretion and bacterial coating also depend on a functional bacterial type IV secretion system, which drives the generation of Th17 cells and the IL-17-dependent expression of the polymeric immunoglobulin receptor PIGR. IgA-/- mice are hypercolonized with C. rodentium in all examined tissues, suffer from more severe weight loss and develop more colitis. In contrast, H. pylori is controlled more efficiently in IgA-/- mice than their WT counterparts. The effects of IgA deficiency of the offspring can be compensated by maternal IgA delivered by WT foster mothers. We attribute the improved immune control observed in IgA-/- mice to IgA-mediated protection from complement killing, as H. pylori colonization is restored to wild type levels in a composite strain lacking both IgA and the central complement component C3. IgA antibodies can thus have protective or detrimental activities depending on the infectious agent.
Collapse
Affiliation(s)
- Mariela Artola-Borán
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Lydia Kirsche
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Angela Fallegger
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Peter Leary
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland; Functional Genomics Center Zürich, University of Zürich/ETHZ, Zürich, Switzerland
| | - Mine Tanriover
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Tanja Goodwin
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Gavin Geiger
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | | | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland; Institute of Biochemistry, Brandenburg Medical School, 16816 Neuruppin, Germany
| | - Isabelle C Arnold
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland; Comprehensive Cancer Center Zürich, Zürich, Switzerland.
| |
Collapse
|
4
|
Vassalakis JA, Yamashita DHS, Midon LM, Cogliati B, Heinemann MB, Amamura TA, Isaac L. Murine C3 of the complement system affects infection by Leptospira interrogans. Microbes Infect 2025; 27:105413. [PMID: 39284496 DOI: 10.1016/j.micinf.2024.105413] [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: 06/12/2024] [Revised: 08/02/2024] [Accepted: 08/30/2024] [Indexed: 03/14/2025]
Abstract
Leptospirosis is an infectious neglected disease estimated to affect more than one million people worldwide each year. The Complement System plays a vital role in eliminating infectious agents. However, its precise role in leptospirosis remains to be fully understood. We investigated the importance of C3 in L. interrogans serovar Kennewicki strain Pomona Fromm (LPF) infection. Lack of C3 leads to decreased leukocyte number, impaired inflammatory response and failure to eliminate bacteria during the early stages of infection, which may cause interstitial nephritis later. These findings could be explained, at least in part, by the lower presence of local opsonins. Furthermore, antibody production against Leptospira was compromised in the absence of C3, highlighting the importance of CR2 in B lymphocyte proliferation and the adjuvant role of C3d in humoral immunity. Leptospires can be eliminated through the urine, and according to our study, the lack of C3 delays the elimination of LPF through urine during the early stages of the infection. These results strongly suggest the crucial role of C3 protein in orchestrating an appropriate inflammatory response against LPF infection and in effectively eliminating the bacteria from the body during the acute phase of leptospirosis.
Collapse
Affiliation(s)
- Julia Avian Vassalakis
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | - Leonardo Moura Midon
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Brazil
| | | | - Thaís Akemi Amamura
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Lourdes Isaac
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
5
|
Kuwazoe H, Sakatani H, Kono M, Saika S, Inoue N, Hotomi M. Complement Component 3 Promotes Regeneration of Olfactory Receptor Neurons. J Transl Med 2025; 105:102200. [PMID: 39581348 DOI: 10.1016/j.labinv.2024.102200] [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: 06/11/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024] Open
Abstract
Olfactory receptor neurons (ORNs) in the olfactory epithelium are characterized by high regenerative capacity even after birth, but the molecular mechanisms involved in ORN regeneration remain unclear. Complement component 3 (C3) has been shown to promote tissue regeneration, so we hypothesized that C3 activates innate immunity and also promotes the regeneration of ORNs. In this study, we investigate the role of C3 in ORN regeneration. We used C3 knockout (KO) and wild-type C57BL/6J mice in this study to examine the olfactory regeneration process for 42 days after methimazole-induced olfactory disorder. To compare the regeneration process after ORN damage between C3 KO and wild-type mice, we conducted olfactory behavioral tests and immunohistologic analysis and examined growth factors and inflammatory cell induction. C3 KO mice showed delayed olfactory recovery with lower olfactory epithelial thickness. In C3 KO mice, ORN maturation was delayed in association with increased accumulation of immature ORNs. In the normal ORN regeneration process, undesirable immature ORNs are produced and eliminated by apoptosis. C3 deficiency reduced neutrophils induced during ORN regeneration, suggesting the involvement of C3 in ORN regeneration through neutrophil-dependent elimination of undesired ORNs. C3 is therefore suggested to have promoted ORN regeneration by preventing the accumulation of immature ORNs. In addition, C3 may assist ORN maturation by participating in ORN axon selection such as synaptic pruning. Our results indicate that C3, which is activated during pathogen infection, also promotes recovery from ORN damage. These findings may lead to new therapeutic strategies for olfactory disorder.
Collapse
Affiliation(s)
- Hiroki Kuwazoe
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Hideki Sakatani
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Masamitsu Kono
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Norimitsu Inoue
- Department of Molecular Genetics, Wakayama Medical University, Wakayama, Japan
| | - Muneki Hotomi
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan.
| |
Collapse
|
6
|
Vandendriessche S, Mattos MS, Bialek EL, Schuermans S, Proost P, Marques PE. Complement activation drives the phagocytosis of necrotic cell debris and resolution of liver injury. Front Immunol 2024; 15:1512470. [PMID: 39759517 PMCID: PMC11696981 DOI: 10.3389/fimmu.2024.1512470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/29/2024] [Indexed: 01/07/2025] Open
Abstract
Cells die by necrosis due to excessive chemical or thermal stress, leading to plasma membrane rupture, release of intracellular components and severe inflammation. The clearance of necrotic cell debris is crucial for tissue recovery and injury resolution, however, the underlying mechanisms are still poorly understood, especially in vivo. This study examined the role of complement proteins in promoting clearance of necrotic cell debris by leukocytes and their influence on liver regeneration. We found that independently of the type of necrotic liver injury, either acetaminophen (APAP) overdose or thermal injury, complement proteins C1q and (i)C3b were deposited specifically on necrotic lesions via the activation of the classical pathway. Importantly, C3 deficiency led to a significant accumulation of necrotic debris and impairment of liver recovery in mice, which was attributed to decreased phagocytosis of debris by recruited neutrophils in vivo. Monocytes and macrophages also took part in debris clearance, although the necessity of C3 and CD11b was dependent on the specific type of necrotic liver injury. Using human neutrophils, we showed that absence of C3 or C1q caused a reduction in the volume of necrotic debris that is phagocytosed, indicating that complement promotes effective debris uptake in mice and humans. Moreover, internalization of opsonized debris induced the expression of pro-resolving genes in a C3-dependent manner, supporting the notion that debris clearance favors the resolution of inflammation. In summary, complement activation at injury sites is a pivotal event for necrotic debris clearance by phagocytes and determinant for efficient recovery from tissue injury.
Collapse
Affiliation(s)
| | | | | | | | | | - Pedro Elias Marques
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
7
|
Yin J, Verschoor A, Yue X, Goldmann T, Heidecke H, Riemekasten G, Petersen F, Yu X. C3 deficiency promotes pulmonary inflammation in AT1R-induced mouse model for systemic sclerosis. Front Immunol 2024; 15:1491324. [PMID: 39737181 PMCID: PMC11683138 DOI: 10.3389/fimmu.2024.1491324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/29/2024] [Indexed: 01/01/2025] Open
Abstract
Introduction Autoantibody-mediated complement activation plays an essential role in a variety of autoimmune disorders. However, the role of complement in systemic sclerosis (SSc) remains largely unknown. In this study, we aimed to determine the role of complement C3 in the development of a recently described SSc mouse model based on autoimmunity to angiotensin II receptor type 1 (AT1R). Methods Mice were immunized with cell membrane extract isolated from Chinese hamster ovary (CHO) cells overexpressing AT1R or non-transfected CHO cells as a control. Peripheral blood, dorsal skin and the lung were then collected to evlauate disease characteristics. Apoptotic cells in the lung of mice were detected using the DeadEnd™ Fluorometric TUNEL System. Results Our results showed that experimental SSc in this model was featured by the deposition of IgG, but not of complement C3, in the lung. After immunization with AT1R, C3-deficient mice developed more severe pulmonary inflammations than wild type controls, whereas skin inflammation and fibrosis were not different as well as the anti-AT1R ab levels. Further, C3-deficient mice showed an increased rate of pulmonary cell apoptosis as compared to controls. The apoptosis rate correlated with the corresponding degree of lung inflammation. Discussion Taken together, our findings suggest an anti-apoptotic and anti-inflammatory role of complement C3 in pulmonary autoimmune inflammation.
Collapse
Affiliation(s)
- Junping Yin
- Priority Area Chronic Lung Diseases, Research Center Borstel - Leibniz Lung Center, Members of the German Center for Lung Research (DZL), Borstel, Germany
| | - Admar Verschoor
- Department of Otorhinolaryngology, Technische Universität München and Klinikum Rechts der Isar, Munich, Germany
- Department of Dermatology, University Clinic of Schleswig Holstein, University of Lübeck, Lübeck, Germany
| | - Xiaoyang Yue
- Priority Area Chronic Lung Diseases, Research Center Borstel - Leibniz Lung Center, Members of the German Center for Lung Research (DZL), Borstel, Germany
- College of Basic Medical Science, Key Laboratory of Basic Research on Regional Diseases, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Medical University, Nanning, China
| | - Torsten Goldmann
- Histology, Research Center Borstel - Leibniz Lung Center, Members of the German Center for Lung Research (DZL), Borstel, Germany
| | | | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, University Clinic of Schleswig Holstein, University of Lübeck, Lübeck, Germany
| | - Frank Petersen
- Priority Area Chronic Lung Diseases, Research Center Borstel - Leibniz Lung Center, Members of the German Center for Lung Research (DZL), Borstel, Germany
| | - Xinhua Yu
- Priority Area Chronic Lung Diseases, Research Center Borstel - Leibniz Lung Center, Members of the German Center for Lung Research (DZL), Borstel, Germany
| |
Collapse
|
8
|
Alic L, Dendinovic K, Papac-Milicevic N. The complement system in lipid-mediated pathologies. Front Immunol 2024; 15:1511886. [PMID: 39635529 PMCID: PMC11614835 DOI: 10.3389/fimmu.2024.1511886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
The complement system, a coordinator and facilitator of the innate immune response, plays an essential role in maintaining host homeostasis. It promotes clearance of pathogen- and danger-associated molecular patterns, regulates adaptive immunity, and can modify various metabolic processes such as energy expenditure, lipid metabolism, and glucose homeostasis. In this review, we will focus on the intricate interplay between complement components and lipid metabolism. More precisely, we will display how alterations in the activation and regulation of the complement system affect pathological outcome in lipid-associated diseases, such as atherosclerosis, obesity, metabolic syndrome, age-related macular degeneration, and metabolic dysfunction-associated steatotic liver disease. In addition to that, we will present and evaluate underlying complement-mediated physiological mechanisms, observed both in vitro and in vivo. Our manuscript will demonstrate the clinical significance of the complement system as a bridging figure between innate immunity and lipid homeostasis.
Collapse
Affiliation(s)
- Lejla Alic
- Department of Medical Biochemistry, Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Kristina Dendinovic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Nikolina Papac-Milicevic
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
Yang Y, Treger RS, Hernandez-Bird J, Lu P, Mao T, Iwasaki A. A B cell screen against endogenous retroviruses identifies glycan-reactive IgM that recognizes a broad array of enveloped viruses. Sci Immunol 2024; 9:eadd6608. [PMID: 39514636 PMCID: PMC11962862 DOI: 10.1126/sciimmunol.add6608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 10/11/2024] [Indexed: 11/16/2024]
Abstract
Endogenous retroviruses (ERVs), comprising a substantial portion of the vertebrate genome, are remnants of ancient genetic invaders. ERVs with near-intact coding potential reactivate in B cell-deficient mice. To study how B cells contribute to host anti-ERV immunity, we used an antigen-baiting strategy to enrich B cells reactive to ERV surface antigens. We identified ERV-reactive B-1 cells expressing germline-encoded natural IgM antibodies in naïve mice, the level of which further increases upon innate immune sensor stimulation. B cell receptor repertoire profiling of ERV-reactive B-1 cells revealed increased usage of the Igh VH gene that gives rise to glycan-specific antibodies targeting terminal N-acetylglucosamine moieties on ERV glycoproteins, which further engage the complement pathway to mediate anti-ERV responses. These same antibodies also recognize glycoproteins of other enveloped viruses but not self-proteins. These results reveal an innate antiviral mechanism of germline-encoded antibodies with broad reactivity to enveloped viruses, which constitutes a natural antibody repertoire capable of preventing the emergence of infectious ERVs.
Collapse
Affiliation(s)
- Yexin Yang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Rebecca S. Treger
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Juan Hernandez-Bird
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Peiwen Lu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Tianyang Mao
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
10
|
Francisco da Silva T, Akemi Amamura T, Cordeiro Valadão I, Carvalho Carneiro M, Morais Freitas V, Paula Lepique A, Isaac L. Complement system component 3 deficiency modulates the phenotypic profile of murine macrophages. Cell Immunol 2024; 405-406:104886. [PMID: 39503081 DOI: 10.1016/j.cellimm.2024.104886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/13/2024] [Accepted: 10/16/2024] [Indexed: 12/02/2024]
Abstract
The Complement System is composed of more than 40 proteins that act in innate and adaptive immunity. C3 is the most abundant one and C3-deficient patients are more susceptible to recurrent and severe infections. Several studies have demonstrated the importance of C3 in controlling infections. However, its role in leukocyte biology is still poorly understood. This study aimed to evaluate several cellular parameters in macrophages from C3-deficient mice and compare them to similar cells from wild-type counterparts. We observed that in the absence of C3, the population of F4/80low macrophages in the peritoneal cavity of thioglycolate-treated mice is diminished, probably due to the lack of chemotactic factors like C3a and low levels of C5a. Using fluorescence microscopy analysis, we observed that macrophages from C3-deficient mice exhibited morphological alterations when compared to similar cells from wild-type mice. We observed a significant increase in the expression of CD11c, which is part of CR4 (CD11c/CD18), in macrophages from C3-deficient compared to cells from wild-type mice. Treatment with 12-o-tetradecanoylphorbol-13-acetate, stimulated ROS production and MAPK activation by macrophages. However, these parameters were lower in macrophages from C3-deficient mice when compared to wild-type counterparts. In addition, the phagocytosis of iC3b-opsonized Zymosan particles was diminished in macrophages from C3-deficient mice. Our results suggest that C3 deficiency in C57Black/6 mice may influence specific morphological and functional parameters of macrophages, cells of fundamental importance for both the innate and acquired immune responses.
Collapse
Affiliation(s)
- Tiago Francisco da Silva
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Thaís Akemi Amamura
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Iuri Cordeiro Valadão
- Tumor Microenvironment Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Milena Carvalho Carneiro
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Vanessa Morais Freitas
- Tumor Microenvironment Laboratory, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Ana Paula Lepique
- Laboratory of Immunomodulation, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Lourdes Isaac
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
11
|
Sedney CJ, Masters J, Callender M, Dewan K, Caulfield A, Harvill ET. Neonatal Neutrophil-mediated Control of Bordetella pertussis Is Disrupted by Pertussis Toxin. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:ji2400363. [PMID: 39475256 PMCID: PMC11605672 DOI: 10.4049/jimmunol.2400363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 10/08/2024] [Indexed: 12/01/2024]
Abstract
The increased susceptibility of infants and young children to some diseases has often been explained as the neonatal immune system (NIS) being incomplete and/or underdeveloped. However, our recent work demonstrated that neonatal mice could clear a Bordetella pertussis (Bp) strain lacking pertussis toxin (PTx) (BpΔptx) much more efficiently than adult mice, indicating that the NIS can be extremely effective, but this ability is highly sensitive to being blocked by PTx. In this article, we investigated immunological mechanisms by which neonates efficiently and rapidly clear BpΔptx to better understand how the NIS functions and how PTx disrupts it. Depleting neutrophils, or blocking their recruitment, inhibited pups' ability to rapidly clear BpΔptx, revealing a critical role for neutrophils. Pups deficient in complement (C3-/-) failed to recruit neutrophils and did not efficiently clear BpΔptx but recovered these abilities upon treatment with C3a. Neutrophil depletion in C3-/- pups led to further failure to control BpΔptx, suggesting that neutrophils and complement have independent roles in rapid clearance of BpΔptx. Depleting or disrupting neutrophils and complement had negligible effect on the rapid growth of wild-type Bp, indicating that PTx blocks these otherwise highly effective aspects of the NIS.
Collapse
Affiliation(s)
- Colleen J. Sedney
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Jillian Masters
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Maiya Callender
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Kalyan Dewan
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Amanda Caulfield
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Eric T. Harvill
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA
| |
Collapse
|
12
|
Murayama MA. Complement C3 deficient mice show more severe imiquimod-induced psoriasiform dermatitis than wild-type mice regardless of the commensal microbiota. Exp Anim 2024; 73:458-467. [PMID: 38945882 PMCID: PMC11534491 DOI: 10.1538/expanim.24-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024] Open
Abstract
The complement active product, C3a, and the receptor C3aR comprise an axis that exerts various biological functions, such as protection against infection. C3a is highly expressed in the inflamed skin and blood from patients with psoriasiform dermatitis. However, the role of the C3a/C3aR axis in psoriasiform dermatitis remains unclear because conflicting results using C3-/- mice have been published. In this study, to elucidate the contribution of commensal microbiota in C3-/- and wild-type (WT) mice were subjected to imiquimod-induced psoriasiform dermatitis under different housing conditions. C3-/- mice showed increased epidermal thickness and keratinocyte proliferation markers in the inflamed ear compared to WT mice upon treatment with IMQ. These inflamed phenotypes were observed in both cohoused and separately housed conditions, and antibiotic treatment did not abolish the aggravation of IMQ-induced psoriasiform dermatitis in C3-/- mice. These results suggested that the difference of commensal microbiota is not important for the C3-involved psoriasiform dermatitis. Keratinocyte hyperproliferation is a major feature of the inflamed skin in patients with psoriasiform dermatitis. In vitro experiments showed that C3a and C3aR agonists inhibited keratinocyte proliferation, which was abolished by introduction of a C3aR antagonist. Collectively, these results suggest that the C3a/C3aR axis plays a critical role in psoriasiform dermatitis development by inhibiting keratinocyte proliferation, regardless of the regulation of the commensal microbiota.
Collapse
Affiliation(s)
- Masanori A Murayama
- Department of Animal Models for Human Diseases, Institute of Biomedical Science, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
13
|
Stark K, Kilani B, Stockhausen S, Busse J, Schubert I, Tran TD, Gaertner F, Leunig A, Pekayvaz K, Nicolai L, Fumagalli V, Stermann J, Stephan F, David C, Müller MB, Heyman B, Lux A, da Palma Guerreiro A, Frenzel LP, Schmidt CQ, Dopler A, Moser M, Chandraratne S, von Brühl ML, Lorenz M, Korff T, Rudelius M, Popp O, Kirchner M, Mertins P, Nimmerjahn F, Iannacone M, Sperandio M, Engelmann B, Verschoor A, Massberg S. Antibodies and complement are key drivers of thrombosis. Immunity 2024; 57:2140-2156.e10. [PMID: 39226900 DOI: 10.1016/j.immuni.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/17/2024] [Accepted: 08/07/2024] [Indexed: 09/05/2024]
Abstract
Venous thromboembolism (VTE) is a common, deadly disease with an increasing incidence despite preventive efforts. Clinical observations have associated elevated antibody concentrations or antibody-based therapies with thrombotic events. However, how antibodies contribute to thrombosis is unknown. Here, we show that reduced blood flow enabled immunoglobulin M (IgM) to bind to FcμR and the polymeric immunoglobulin receptor (pIgR), initiating endothelial activation and platelet recruitment. Subsequently, the procoagulant surface of activated platelets accommodated antigen- and FcγR-independent IgG deposition. This leads to classical complement activation, setting in motion a prothrombotic vicious circle. Key elements of this mechanism were present in humans in the setting of venous stasis as well as in the dysregulated immunothrombosis of COVID-19. This antibody-driven thrombosis can be prevented by pharmacologically targeting complement. Hence, our results uncover antibodies as previously unrecognized central regulators of thrombosis. These findings carry relevance for therapeutic application of antibodies and open innovative avenues to target thrombosis without compromising hemostasis.
Collapse
Affiliation(s)
- Konstantin Stark
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.
| | - Badr Kilani
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Sven Stockhausen
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Johanna Busse
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Irene Schubert
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Thuy-Duong Tran
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Florian Gaertner
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany; Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Alexander Leunig
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Kami Pekayvaz
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Leo Nicolai
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Valeria Fumagalli
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Julia Stermann
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Felix Stephan
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Christian David
- Institute for Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine, Biomedical Center (BMC) LMU Munich, Munich, Germany
| | - Martin B Müller
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany; Department of Anaesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Birgitta Heyman
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Anja Lux
- Department of Biology, Institute of Genetics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Medical Immunology Campus Erlangen (MICE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Alexandra da Palma Guerreiro
- Department I of Internal Medicine, University Hospital Cologne, Cologne 50937, Germany; Center of Integrated Oncology ABCD, University Hospital of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50937, Germany
| | - Lukas P Frenzel
- Department I of Internal Medicine, University Hospital Cologne, Cologne 50937, Germany; Center of Integrated Oncology ABCD, University Hospital of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50937, Germany
| | - Christoph Q Schmidt
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Center, Ulm, Germany
| | - Arthur Dopler
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Center, Ulm, Germany
| | - Markus Moser
- Department of Molecular Medicine, Max-Planck-Institute of Biochemistry, Martinsried, Germany; Institute of Experimental Hematology, TranslaTUM, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Sue Chandraratne
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Marie-Luise von Brühl
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Michael Lorenz
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Thomas Korff
- Division of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Martina Rudelius
- Institute of Pathology, Ludwig-Maximilian University, Munich, Germany
| | - Oliver Popp
- Max Delbrück Center for Molecular Medicine (MDC) and Berlin Institute of Health (BIH), Berlin, Germany; German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Marieluise Kirchner
- Max Delbrück Center for Molecular Medicine (MDC) and Berlin Institute of Health (BIH), Berlin, Germany; German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Philipp Mertins
- Max Delbrück Center for Molecular Medicine (MDC) and Berlin Institute of Health (BIH), Berlin, Germany; German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Falk Nimmerjahn
- Department of Biology, Institute of Genetics, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Medical Immunology Campus Erlangen (MICE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Matteo Iannacone
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Markus Sperandio
- Institute for Cardiovascular Physiology and Pathophysiology, Walter Brendel Center for Experimental Medicine, Biomedical Center (BMC) LMU Munich, Munich, Germany
| | - Bernd Engelmann
- Institut für Laboratoriumsmedizin, University Hospital, LMU Munich, Munich, Germany
| | - Admar Verschoor
- Department of Dermatology, Allergy, and Venereology, University of Lübeck, Lübeck, Germany; Department of Otorhinolaryngology, Technische Universität München and Klinikum Rechts der Isar, Munich, Germany.
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, University Hospital, LMU Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany; Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| |
Collapse
|
14
|
Schmidt M, Linder AT, Korn M, Schellenberg N, Meyer SJ, Nimmerjahn F, Werner A, Abeln M, Gerardy-Schahn R, Münster-Kühnel AK, Nitschke L. Sialic acids on T cells are crucial for their maintenance and survival. Front Immunol 2024; 15:1359494. [PMID: 38947328 PMCID: PMC11211268 DOI: 10.3389/fimmu.2024.1359494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/27/2024] [Indexed: 07/02/2024] Open
Abstract
Sialic acids are found as terminal sugars on glycan structures on cellular surfaces. T cells carry these sialoglycans abundantly, and they are thought to serve multiple functions in cell adhesion, cell migration, and protection from complement attack. We studied the role of sialoglycans on T cells in a mouse model with a T cell-specific deletion of cytidine monophosphate-sialic acid synthase (CMAS), the enzyme that is crucial for the synthesis of sialoglycans. These mice showed a T-cell deficiency in peripheral lymphoid organs. Many T cells with an undeleted Cmas allele were found in the periphery, suggesting that they escaped the Cre-mediated deletion. The remaining peripheral T cells of T cell-specific Cmas KO mice had a memory-like phenotype. Additional depletion of the complement factor C3 could not rescue the phenotype, showing that the T-cell defect was not caused by a host complement activity. Cmas-deficient T cells showed a high level of activated caspase 3, indicating an ongoing apoptosis. In bone marrow chimeric cellular transfer experiments, we observed a strong competitive disadvantage of Cmas-deficient T cells compared to wild-type T cells. These results show that sialoglycans on the surface of T cells are crucial for T-cell survival and maintenance. This function has not been recognized before and is similar to the function of sialoglycans on B cells.
Collapse
Affiliation(s)
- Michael Schmidt
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Alexandra T. Linder
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Marina Korn
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Nick Schellenberg
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Sarah J. Meyer
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Falk Nimmerjahn
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Anja Werner
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| | - Markus Abeln
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Rita Gerardy-Schahn
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | | | - Lars Nitschke
- Division of Genetics, Department of Biology, University of Erlangen, Erlangen, Germany
| |
Collapse
|
15
|
Bin S, Yoo M, Molinari P, Gentile M, Budge K, Cantarelli C, Khan Y, La Manna G, Baldwin WM, Dvorina N, Cravedi P, Gusella GL. Reduced decay-accelerating factor expression promotes complement-mediated cystogenesis in murine ADPKD. JCI Insight 2024; 9:e175220. [PMID: 38912583 PMCID: PMC11383362 DOI: 10.1172/jci.insight.175220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 05/15/2024] [Indexed: 06/25/2024] Open
Abstract
Patients with autosomal dominant polycystic kidney disease (ADPKD), a genetic disease due to mutations of the PKD1 or PKD2 gene, show signs of complement activation in the urine and cystic fluid, but their pathogenic role in cystogenesis is unclear. We tested the causal relationship between complement activation and cyst growth using a Pkd1KO renal tubular cell line and newly generated conditional Pkd1-/- C3-/- mice. Pkd1-deficient tubular cells have increased expression of complement-related genes (C3, C5, CfB, C3ar, and C5ar1), while the gene and protein expression of complement regulators DAF, CD59, and Crry is decreased. Pkd1-/- C3-/- mice are unable to fully activate the complement cascade and are characterized by a significantly slower kidney cystogenesis, preserved renal function, and reduced intrarenal inflammation compared with Pkd1-/- C3+/+ controls. Transgenic expression of the cytoplasmic C-terminal tail of Pkd1 in Pkd1KO cells lowered C5ar1 expression, restored Daf levels, and reduced cell proliferation. Consistently, both DAF overexpression and pharmacological inhibition of C5aR1 (but not C3aR) reduced Pkd1KO cell proliferation. In conclusion, the loss of Pkd1 promotes unleashed activation of locally produced complement by downregulating DAF expression in renal tubular cells. Increased C5a formation and C5aR1 activation in tubular cells promotes cyst growth, offering a new therapeutic target.
Collapse
Affiliation(s)
- Sofia Bin
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero- University of Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Miran Yoo
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Paolo Molinari
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Micaela Gentile
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Unità Operativa Nefrologia, Azienda-Ospedaliero University of Parma, Department of Medicine and Syrgery, University of Parma, Italy
| | - Kelly Budge
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Chiara Cantarelli
- Unità Operativa Nefrologia, Azienda-Ospedaliero University of Parma, Department of Medicine and Syrgery, University of Parma, Italy
| | - Yaseen Khan
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gaetano La Manna
- Nephrology, Dialysis and Kidney Transplant Unit, IRCCS Azienda Ospedaliero- University of Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - William M Baldwin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nina Dvorina
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Paolo Cravedi
- Translational Transplant Research Center and Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - G Luca Gusella
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
16
|
Melchior K, Gerner RR, Hossain S, Nuccio SP, Moreira CG, Raffatellu M. IL-22-dependent responses and their role during Citrobacter rodentium infection. Infect Immun 2024; 92:e0009924. [PMID: 38557196 PMCID: PMC11075456 DOI: 10.1128/iai.00099-24] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
The mouse pathogen Citrobacter rodentium is utilized as a model organism for studying infections caused by the human pathogens enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC) and to elucidate mechanisms of mucosal immunity. In response to C. rodentium infection, innate lymphoid cells and T cells secrete interleukin (IL)-22, a cytokine that promotes mucosal barrier function. IL-22 plays a pivotal role in enabling mice to survive and recover from C. rodentium infection, although the exact mechanisms involved remain incompletely understood. Here, we investigated whether particular components of the host response downstream of IL-22 contribute to the cytokine's protective effects during C. rodentium infection. In line with previous research, mice lacking the IL-22 gene (Il22-/- mice) were highly susceptible to C. rodentium infection. To elucidate the role of specific antimicrobial proteins modulated by IL-22, we infected the following knockout mice: S100A9-/- (calprotectin), Lcn2-/- (lipocalin-2), Reg3b-/- (Reg3β), Reg3g-/- (Reg3γ), and C3-/- (C3). All knockout mice tested displayed a considerable level of resistance to C. rodentium infection, and none phenocopied the lethality observed in Il22-/- mice. By investigating another arm of the IL-22 response, we observed that C. rodentium-infected Il22-/- mice exhibited an overall decrease in gene expression related to intestinal barrier integrity as well as significantly elevated colonic inflammation, gut permeability, and pathogen levels in the spleen. Taken together, these results indicate that host resistance to lethal C. rodentium infection may depend on multiple antimicrobial responses acting in concert, or that other IL-22-regulated processes, such as tissue repair and maintenance of epithelial integrity, play crucial roles in host defense to attaching and effacing pathogens.
Collapse
Affiliation(s)
- Karine Melchior
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California, USA
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Romana R. Gerner
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California, USA
- School of Life Sciences, ZIEL – Institute for Food and Health, Freising-Weihenstephan, Technical University of Munich, Munich, Germany
- Department of Internal Medicine III, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Suzana Hossain
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Sean-Paul Nuccio
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Cristiano Gallina Moreira
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Manuela Raffatellu
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines (CU-UCSD cMAV), La Jolla, California, USA
| |
Collapse
|
17
|
Wang X, Gong Q, Nie H, Tu J, Fan W, Tan X. High level of C3 is associated with Th2 immune response and liver fibrosis in patients with schistosomiasis. Parasite Immunol 2024; 46:e13029. [PMID: 38465509 DOI: 10.1111/pim.13029] [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: 09/20/2023] [Revised: 01/10/2024] [Accepted: 02/05/2024] [Indexed: 03/12/2024]
Abstract
Long-term infection of schistosomiasis will seriously affect the liver health of patients. The serum of 334 chronic Schistosoma japonicum patients and 149 healthy volunteers was collected. Compared with heathy people, the level of C4 (complement 4) was increased, and the level of C3 (complement 3) was in an obvious skewed distribution. ELISA was performed to detect the serum cytokines, the results showed that the levels of IFN-γ (interferon-γ), IL (interleukin)-2 and TNF-α (tumour necrosis factor-α) were reduced, while the levels of Th2 cytokines (IL-4, IL-6 and IL-10) were increased. In the serum of patients with high C3, the secretion of HA (hyaluronic acid), LN (laminin), IV-C (type IV collagen) and PCIII (type III procollagen) were increased, the activation of hepatic stellate cells was promoted. Exogenous human recombinant C3 made mice liver structure of the mice damaged and collagen deposition. IFN-γ and IFN-γ/IL-4 were decreased, while HA, LN, PCIII and IV-C were increased, and the expressions of α-SMA and TGF-β1 in liver tissues were up-regulated. However, the addition of IFN-γ partially reversed the effect of C3 on promoting fibrosis. High level of C3 is associated with Th2 immune response and liver fibrosis in patients with schistosomiasis.
Collapse
Affiliation(s)
- Xianmo Wang
- Clinical Laboratory, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei Province, China
| | - Quan Gong
- Yangtze University, Jingzhou, Hubei Province, China
| | - Hao Nie
- Yangtze University, Jingzhou, Hubei Province, China
| | - Jiancheng Tu
- Clinical Laboratory, The Second Clinical College of Wuhan University, Wuhan, Hubei province, China
| | - Wen Fan
- Clinical Laboratory, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei Province, China
| | - Xiaoping Tan
- Gastroenterology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei Province, China
| |
Collapse
|
18
|
Xiao MT, Ellsworth CR, Qin X. Emerging role of complement in COVID-19 and other respiratory virus diseases. Cell Mol Life Sci 2024; 81:94. [PMID: 38368584 PMCID: PMC10874912 DOI: 10.1007/s00018-024-05157-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/03/2024] [Accepted: 02/03/2024] [Indexed: 02/19/2024]
Abstract
The complement system, a key component of innate immunity, provides the first line of defense against bacterial infection; however, the COVID-19 pandemic has revealed that it may also engender severe complications in the context of viral respiratory disease. Here, we review the mechanisms of complement activation and regulation and explore their roles in both protecting against infection and exacerbating disease. We discuss emerging evidence related to complement-targeted therapeutics in COVID-19 and compare the role of the complement in other respiratory viral diseases like influenza and respiratory syncytial virus. We review recent mechanistic studies and animal models that can be used for further investigation. Novel knockout studies are proposed to better understand the nuances of the activation of the complement system in respiratory viral diseases.
Collapse
Affiliation(s)
- Mark T Xiao
- Division of Comparative Pathology, Tulane National Primate Research Center, Health Sciences Campus, 18703 Three Rivers Road, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Calder R Ellsworth
- Division of Comparative Pathology, Tulane National Primate Research Center, Health Sciences Campus, 18703 Three Rivers Road, Covington, LA, 70433, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Xuebin Qin
- Division of Comparative Pathology, Tulane National Primate Research Center, Health Sciences Campus, 18703 Three Rivers Road, Covington, LA, 70433, USA.
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
19
|
Pizzato HA, Alonso-Guallart P, Woods J, Connelly JP, Fehniger TA, Atkinson JP, Pruett-Miller SM, Monsma FJ, Bhattacharya D. Engineering human pluripotent stem cell lines to evade xenogeneic transplantation barriers. Stem Cell Reports 2024; 19:299-313. [PMID: 38215755 PMCID: PMC10874864 DOI: 10.1016/j.stemcr.2023.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 01/14/2024] Open
Abstract
Successful allogeneic human pluripotent stem cell (hPSC)-derived therapies must overcome immunological rejection by the recipient. To build reagents to define these barriers, we genetically ablated β2M, TAP1, CIITA, CD74, MICA, and MICB to limit expression of HLA-I, HLA-II, and natural killer (NK) cell activating ligands in hPSCs. Transplantation of these cells that also expressed covalent single chain trimers of Qa1 and H2-Kb to inhibit NK cells and CD55, Crry, and CD59 to inhibit complement deposition led to persistent teratomas in wild-type mice. Transplantation of HLA-deficient hPSCs into mice genetically deficient in complement and depleted of NK cells also led to persistent teratomas. Thus, T cell, NK cell, and complement evasion are necessary to prevent immunological rejection of hPSCs and their progeny. These cells and versions expressing human orthologs of immune evasion factors can be used to define cell type-specific immune barriers and conduct preclinical testing in immunocompetent mouse models.
Collapse
Affiliation(s)
- Hannah A Pizzato
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | | | - James Woods
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Jon P Connelly
- Department of Cell & Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Todd A Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John P Atkinson
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shondra M Pruett-Miller
- Department of Cell & Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Frederick J Monsma
- The New York Stem Cell Foundation Research Institute, New York, NY 10019, USA
| | - Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ 85724, USA; Department of Surgery, University of Arizona College of Medicine, Tucson, AZ 85724, USA; BIO5 Institute, University of Arizona, Tucson, AZ 85724, USA.
| |
Collapse
|
20
|
Alghory A, Alwani A, Seida R, Alrais M, Alwani A, Mahroum N. Streptococcus pneumoniae and Autoimmunity. INFECTION AND AUTOIMMUNITY 2024:449-458. [DOI: 10.1016/b978-0-323-99130-8.00050-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
21
|
Dahmani M, Zhu JC, Cook JH, Riley SP. Anaphylatoxin signaling activates macrophages to control intracellular Rickettsia proliferation. Microbiol Spectr 2023; 11:e0253823. [PMID: 37855623 PMCID: PMC10714731 DOI: 10.1128/spectrum.02538-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/11/2023] [Indexed: 10/20/2023] Open
Abstract
IMPORTANCE Pathogenic Rickettsia species are extremely dangerous bacteria that grow within the cytoplasm of host mammalian cells. In most cases, these bacteria are able to overpower the host cell and grow within the protected environment of the cytoplasm. However, a dramatic conflict occurs when Rickettsia encounter innate immune cells; the bacteria can "win" by taking over the host, or the bacteria can "lose" if the host cell efficiently fights the infection. This manuscript examines how the immune complement system is able to detect the presence of Rickettsia and alert nearby cells. Byproducts of complement activation called anaphylatoxins are signals that "activate" innate immune cells to mount an aggressive defensive strategy. This study enhances our collective understanding of the innate immune reaction to intracellular bacteria and will contribute to future efforts at controlling these dangerous infections.
Collapse
Affiliation(s)
- Mustapha Dahmani
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA
| | - Jinyi C. Zhu
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA
| | - Jack H. Cook
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA
| | - Sean P. Riley
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA
- Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| |
Collapse
|
22
|
Nishioka T, Katayama KI, Kumegawa S, Isono K, Baba T, Tsujimoto H, Yamada G, Inoue N, Asamura S. Increased infiltration of CD4 + T cell in the complement deficient lymphedema model. BMC Immunol 2023; 24:42. [PMID: 37940849 PMCID: PMC10633916 DOI: 10.1186/s12865-023-00580-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 10/25/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Lymphedema is an intractable disease that can be caused by injury to lymphatic vessels, such as by surgical treatments for cancer. It can lead to impaired joint mobility in the extremities and reduced quality of life. Chronic inflammation due to infiltration of various immune cells in an area of lymphedema is thought to lead to local fibrosis, but the molecular pathogenesis of lymphedema remains unclear. Development of effective therapies requires elucidation of the immunological mechanisms involved in the progression of lymphedema. The complement system is part of the innate immune system which has a central role in the elimination of invading microbes and acts as a scavenger of altered host cells, such as apoptotic and necrotic cells and cellular debris. Complement-targeted therapies have recently been clinically applied to various diseases caused by complement overactivation. In this context, we aimed to determine whether complement activation is involved in the development of lymphedema. RESULTS Our mouse tail lymphedema models showed increased expression of C3, and that the classical or lectin pathway was locally activated. Complement activation was suggested to be involved in the progression of lymphedema. In comparison of the C3 knockout (KO) mouse lymphedema model and wild-type mice, there was no difference in the degree of edema at three weeks postoperatively, but the C3 KO mice had a significant increase of TUNEL+ necrotic cells and CD4+ T cells. Infiltration of macrophages and granulocytes was not significantly elevated in C3 KO or C5 KO mice compared with in wild-type mice. Impaired opsonization and decreased migration of macrophages and granulocytes due to C3 deficiency should therefore induce the accumulation of dead cells and may lead to increased infiltration of CD4+ T cells. CONCLUSIONS Vigilance for exacerbation of lymphedema is necessary when surgical treatments have the potential to injure lymphatic vessels in patients undergoing complement-targeted therapies or with complement deficiency. Future studies should aim to elucidate the molecular mechanism of CD4+ T cell infiltration by accumulated dead cells.
Collapse
Affiliation(s)
- Toshihiko Nishioka
- Department of Plastic Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
- Department of Molecular Genetics, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Kei-Ichi Katayama
- Department of Molecular Genetics, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Shinji Kumegawa
- Department of Plastic Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Kyoichi Isono
- Laboratory Animal Center, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Takashi Baba
- Department of Molecular Genetics, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Hiroshi Tsujimoto
- Department of Molecular Genetics, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Gen Yamada
- Department of Plastic Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Norimitsu Inoue
- Department of Molecular Genetics, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan.
| | - Shinichi Asamura
- Department of Plastic Surgery, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| |
Collapse
|
23
|
Mendiola AS, Yan Z, Dixit K, Johnson JR, Bouhaddou M, Meyer-Franke A, Shin MG, Yong Y, Agrawal A, MacDonald E, Muthukumar G, Pearce C, Arun N, Cabriga B, Meza-Acevedo R, Alzamora MDPS, Zamvil SS, Pico AR, Ryu JK, Krogan NJ, Akassoglou K. Defining blood-induced microglia functions in neurodegeneration through multiomic profiling. Nat Immunol 2023; 24:1173-1187. [PMID: 37291385 PMCID: PMC10307624 DOI: 10.1038/s41590-023-01522-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 04/24/2023] [Indexed: 06/10/2023]
Abstract
Blood protein extravasation through a disrupted blood-brain barrier and innate immune activation are hallmarks of neurological diseases and emerging therapeutic targets. However, how blood proteins polarize innate immune cells remains largely unknown. Here, we established an unbiased blood-innate immunity multiomic and genetic loss-of-function pipeline to define the transcriptome and global phosphoproteome of blood-induced innate immune polarization and its role in microglia neurotoxicity. Blood induced widespread microglial transcriptional changes, including changes involving oxidative stress and neurodegenerative genes. Comparative functional multiomics showed that blood proteins induce distinct receptor-mediated transcriptional programs in microglia and macrophages, such as redox, type I interferon and lymphocyte recruitment. Deletion of the blood coagulation factor fibrinogen largely reversed blood-induced microglia neurodegenerative signatures. Genetic elimination of the fibrinogen-binding motif to CD11b in Alzheimer's disease mice reduced microglial lipid metabolism and neurodegenerative signatures that were shared with autoimmune-driven neuroinflammation in multiple sclerosis mice. Our data provide an interactive resource for investigation of the immunology of blood proteins that could support therapeutic targeting of microglia activation by immune and vascular signals.
Collapse
Affiliation(s)
- Andrew S Mendiola
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Zhaoqi Yan
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Karuna Dixit
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | | | - Mehdi Bouhaddou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA
| | | | | | - Yu Yong
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | | | - Eilidh MacDonald
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | | | - Clairice Pearce
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Nikhita Arun
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Belinda Cabriga
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Rosa Meza-Acevedo
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Maria Del Pilar S Alzamora
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
| | - Scott S Zamvil
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | | | - Jae Kyu Ryu
- Gladstone Institutes, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Nevan J Krogan
- Gladstone Institutes, San Francisco, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
| | - Katerina Akassoglou
- Gladstone Institutes, San Francisco, CA, USA.
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA.
| |
Collapse
|
24
|
Pizzato HA, Alonso-Guallart P, Woods J, Johannesson B, Connelly JP, Fehniger TA, Atkinson JP, Pruett-Miller SM, Monsma FJ, Bhattacharya D. Engineering Human Pluripotent Stem Cell Lines to Evade Xenogeneic Transplantation Barriers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.27.546594. [PMID: 37425790 PMCID: PMC10326974 DOI: 10.1101/2023.06.27.546594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Allogeneic human pluripotent stem cell (hPSC)-derived cells and tissues for therapeutic transplantation must necessarily overcome immunological rejection by the recipient. To define these barriers and to create cells capable of evading rejection for preclinical testing in immunocompetent mouse models, we genetically ablated β2m, Tap1, Ciita, Cd74, Mica, and Micb to limit expression of HLA-I, HLA-II, and natural killer cell activating ligands in hPSCs. Though these and even unedited hPSCs readily formed teratomas in cord blood-humanized immunodeficient mice, grafts were rapidly rejected by immunocompetent wild-type mice. Transplantation of these cells that also expressed covalent single chain trimers of Qa1 and H2-Kb to inhibit natural killer cells and CD55, Crry, and CD59 to inhibit complement deposition led to persistent teratomas in wild-type mice. Expression of additional inhibitory factors such as CD24, CD47, and/or PD-L1 had no discernible impact on teratoma growth or persistence. Transplantation of HLA-deficient hPSCs into mice genetically deficient in complement and depleted of natural killer cells also led to persistent teratomas. Thus, T cell, NK cell, and complement evasion are necessary to prevent immunological rejection of hPSCs and their progeny. These cells and versions expressing human orthologs of immune evasion factors can be used to refine tissue- and cell type-specific immune barriers, and to conduct preclinical testing in immunocompetent mouse models.
Collapse
Affiliation(s)
- Hannah A. Pizzato
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | | | - James Woods
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | | | - Jon P. Connelly
- Department of Cell & Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
- Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Todd A. Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - John P. Atkinson
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Shondra M. Pruett-Miller
- Department of Cell & Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
- Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | | | - Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
25
|
Sedney CJ, Harvill ET. The Neonatal Immune System and Respiratory Pathogens. Microorganisms 2023; 11:1597. [PMID: 37375099 PMCID: PMC10301501 DOI: 10.3390/microorganisms11061597] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/02/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Neonates are more susceptible to some pathogens, particularly those that cause infection in the respiratory tract. This is often attributed to an incompletely developed immune system, but recent work demonstrates effective neonatal immune responses to some infection. The emerging view is that neonates have a distinctly different immune response that is well-adapted to deal with unique immunological challenges of the transition from a relatively sterile uterus to a microbe-rich world, tending to suppress potentially dangerous inflammatory responses. Problematically, few animal models allow a mechanistic examination of the roles and effects of various immune functions in this critical transition period. This limits our understanding of neonatal immunity, and therefore our ability to rationally design and develop vaccines and therapeutics to best protect newborns. This review summarizes what is known of the neonatal immune system, focusing on protection against respiratory pathogens and describes challenges of various animal models. Highlighting recent advances in the mouse model, we identify knowledge gaps to be addressed.
Collapse
Affiliation(s)
| | - Eric T. Harvill
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
26
|
Pendse M, De Selle H, Vo N, Quinn G, Dende C, Li Y, Salinas CN, Srinivasan T, Propheter DC, Crofts AA, Koo E, Hassell B, Ruhn KA, Raj P, Obata Y, Hooper LV. Macrophages regulate gastrointestinal motility through complement component 1q. eLife 2023; 12:e78558. [PMID: 37159507 PMCID: PMC10185340 DOI: 10.7554/elife.78558] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/17/2023] [Indexed: 05/11/2023] Open
Abstract
Peristaltic movement of the intestine propels food down the length of the gastrointestinal tract to promote nutrient absorption. Interactions between intestinal macrophages and the enteric nervous system regulate gastrointestinal motility, yet we have an incomplete understanding of the molecular mediators of this crosstalk. Here, we identify complement component 1q (C1q) as a macrophage product that regulates gut motility. Macrophages were the predominant source of C1q in the mouse intestine and most extraintestinal tissues. Although C1q mediates the complement-mediated killing of bacteria in the bloodstream, we found that C1q was not essential for the immune defense of the intestine. Instead, C1q-expressing macrophages were located in the intestinal submucosal and myenteric plexuses where they were closely associated with enteric neurons and expressed surface markers characteristic of nerve-adjacent macrophages in other tissues. Mice with a macrophage-specific deletion of C1qa showed changes in enteric neuronal gene expression, increased neurogenic activity of peristalsis, and accelerated intestinal transit. Our findings identify C1q as a key regulator of gastrointestinal motility and provide enhanced insight into the crosstalk between macrophages and the enteric nervous system.
Collapse
Affiliation(s)
- Mihir Pendse
- Department of Immunology, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Haley De Selle
- Department of Immunology, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Nguyen Vo
- Department of Immunology, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Gabriella Quinn
- Department of Immunology, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Chaitanya Dende
- Department of Immunology, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Yun Li
- Department of Immunology, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Cristine N Salinas
- Department of Immunology, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Tarun Srinivasan
- Department of Immunology, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Daniel C Propheter
- Department of Immunology, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Alexander A Crofts
- Department of Immunology, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Eugene Koo
- Department of Immunology, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Brian Hassell
- Department of Immunology, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Kelly A Ruhn
- Department of Immunology, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Prithvi Raj
- Department of Immunology, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Yuuki Obata
- Department of Immunology, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Lora V Hooper
- Department of Immunology, The University of Texas Southwestern Medical CenterDallasUnited States
- The Howard Hughes Medical Institute, The University of Texas Southwestern Medical CenterDallasUnited States
| |
Collapse
|
27
|
Zhou J, Qiao ML, Jahejo AR, Han XY, Wang P, Wang Y, Ren JL, Niu S, Zhao YJ, Zhang D, Bi YH, Wang QH, Si LL, Fan RW, Shang GJ, Tian WX. Effect of Avian Influenza Virus subtype H9N2 on the expression of complement-associated genes in chicken erythrocytes. Br Poult Sci 2023:1-9. [PMID: 36939295 DOI: 10.1080/00071668.2023.2191308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
The H9N2 subtype avian influenza virus can infect both chickens and humans. Previous studies have reported a role for erythrocytes in immunity. However, the role of H9N2 against chicken erythrocytes and the presence of complement-related genes in erythrocytes has not been studied. This research investigated the effect of H9N2 on complement-associated gene expression in chicken erythrocytes. The expression of complement-associated genes (C1s, C1q, C2, C3, C3ar1, C4, C4a, C5, C5ar1, C7, CD93 and CFD) was detected by reverse transcription-polymerase chain reaction (RT-PCR). Quantitative Real-Time PCR (qRT-PCR) was used to analyse the differential expression of complement-associated genes in chicken erythrocytes at 0 h, 2 h, 6 h and 10 h after the interaction between H9N2 virus and chicken erythrocytes in vitro and 3, 7 and 14 d after H9N2 virus nasal infection of chicks. Expression levels of C1q, C4, C1s, C2, C3, C5, C7 and CD93 were significantly up-regulated at 2 h and significantly down-regulated at 10 h. Gene expression levels of C1q, C3ar1, C4a, CFD and C5ar1 were seen to be different at each time point. The expression levels of C1q, C4, C1s, C2, C3, C5, C7, CFD, C3ar1, C4a and C5ar1 were significantly up-regulated at 7 d and the gene expression of levels of C3, CD93 and C5ar1 were seen to be different at each time point. The results confirmed that all the complement-associated genes were expressed in chicken erythrocytes and showed the H9N2 virus interaction with chicken erythrocytes and subsequent regulation of chicken erythrocyte complement-associated genes expression. This study reported, for the first time, the relationship between H9N2 and complement system of chicken erythrocytes, which will provide a foundation for further research into the prevention and control of H9N2 infection.
Collapse
Affiliation(s)
- J Zhou
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - M L Qiao
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - A R Jahejo
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - X Y Han
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - P Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Y Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - J L Ren
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - S Niu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Y J Zhao
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - D Zhang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Y H Bi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, Institute of Microbiology, Center for Influenza Research and Early-warning (CASCIRE), Chinese Academy of Sciences, Beijing, China
| | - Q H Wang
- CAS Key Laboratory of Microbial Physiological and Metabolic Engineering, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - L L Si
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - R W Fan
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - G J Shang
- Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - W X Tian
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,Shanxi Key Laboratory of protein structure determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| |
Collapse
|
28
|
Gnanaguru G, Tabor SJ, Bonilla GM, Sadreyev R, Yuda K, Köhl J, Connor KM. Microglia refine developing retinal astrocytic and vascular networks through the complement C3/C3aR axis. Development 2023; 150:dev201047. [PMID: 36762625 PMCID: PMC10110418 DOI: 10.1242/dev.201047] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023]
Abstract
Microglia, a resident immune cell of the central nervous system (CNS), play a pivotal role in facilitating neurovascular development through mechanisms that are not fully understood. Previous reports indicate a role for microglia in regulating astrocyte density. This current work resolves the mechanism through which microglia facilitate astrocyte spatial patterning and superficial vascular bed formation in the neuroretina during development. Ablation of microglia increased astrocyte density and altered spatial patterning. Mechanistically, we show that microglia regulate the formation of the spatially organized astrocyte template required for subsequent vascular growth, through the complement C3/C3aR axis during neuroretinal development. Lack of C3 or C3aR hindered the developmental phagocytic removal of astrocyte bodies and resulted in increased astrocyte density. In addition, increased astrocyte density was associated with elevated proangiogenic extracellular matrix gene expression in C3- and C3aR-deficient retinas, resulting in increased vascular density. These data demonstrate that microglia regulate developmental astrocyte and vascular network spatial patterning in the neuroretina via the complement axis.
Collapse
Affiliation(s)
- Gopalan Gnanaguru
- Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Steven J. Tabor
- Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Gracia M. Bonilla
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ruslan Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Kentaro Yuda
- Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Kip M. Connor
- Angiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
29
|
Wang J, Chen HS, Li HH, Wang HJ, Zou RS, Lu XJ, Wang J, Nie BB, Wu JF, Li S, Shan BC, Wu PF, Long LH, Hu ZL, Chen JG, Wang F. Microglia-dependent excessive synaptic pruning leads to cortical underconnectivity and behavioral abnormality following chronic social defeat stress in mice. Brain Behav Immun 2023; 109:23-36. [PMID: 36581303 DOI: 10.1016/j.bbi.2022.12.019] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/17/2022] [Accepted: 12/24/2022] [Indexed: 12/28/2022] Open
Abstract
Synapse loss in medial prefrontal cortex (mPFC) has been implicated in stress-related mood disorders, such as depression. However, the exact effect of synapse elimination in the depression and how it is triggered are largely unknown. Through repeated longitudinal imaging of mPFC in the living brain, we found both presynaptic and postsynaptic components were declined, together with the impairment of synapse remodeling and cross-synaptic signal transmission in the mPFC during chronic stress. Meanwhile, chronic stress also induced excessive microglia phagocytosis, leading to engulfment of excitatory synapses. Further investigation revealed that the elevated complement C3 during the stress acted as the tag of synapses to be eliminated by microglia. Besides, chronic stress induced a reduction of the connectivity between the mPFC and neighbor regions. C3 knockout mice displayed significant reduction of synaptic pruning and alleviation of disrupted functional connectivity in mPFC, resulting in more resilience to chronic stress. These results indicate that complement-mediated excessive microglia phagocytosis in adulthood induces synaptic dysfunction and cortical hypo-connectivity, leading to stress-related behavioral abnormality.
Collapse
Affiliation(s)
- Ji Wang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei 430030, China
| | - Hong-Sheng Chen
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei 430030, China
| | - Hou-Hong Li
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei 430030, China
| | - Hua-Jie Wang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei 430030, China
| | - Ruo-Si Zou
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei 430030, China
| | - Xiao-Jia Lu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei 430030, China
| | - Jie Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bin-Bin Nie
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin-Feng Wu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuang Li
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bao-Ci Shan
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Key Laboratory of Magnetic Resonance in Biological Systems, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peng-Fei Wu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei 430030, China; The Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, 430030 Wuhan, China; Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Wuhan City, Hubei 430030, China; Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Li-Hong Long
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei 430030, China; The Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, 430030 Wuhan, China; Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Wuhan City, Hubei 430030, China; Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Zhuang-Li Hu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei 430030, China; The Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, 430030 Wuhan, China; Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Wuhan City, Hubei 430030, China; Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, 430030 Wuhan, China.
| | - Jian-Guo Chen
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei 430030, China; The Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, 430030 Wuhan, China; Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Wuhan City, Hubei 430030, China; Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, 430030 Wuhan, China.
| | - Fang Wang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, Hubei 430030, China; The Research Center for Depression, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, 430030 Wuhan, China; Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Wuhan City, Hubei 430030, China; Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, 430030 Wuhan, China.
| |
Collapse
|
30
|
Gibson BG, Cox TE, Marchbank KJ. Contribution of animal models to the mechanistic understanding of Alternative Pathway and Amplification Loop (AP/AL)-driven Complement-mediated Diseases. Immunol Rev 2023; 313:194-216. [PMID: 36203396 PMCID: PMC10092198 DOI: 10.1111/imr.13141] [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] [Indexed: 01/20/2023]
Abstract
This review aimed to capture the key findings that animal models have provided around the role of the alternative pathway and amplification loop (AP/AL) in disease. Animal models, particularly mouse models, have been incredibly useful to define the role of complement and the alternative pathway in health and disease; for instance, the use of cobra venom factor and depletion of C3 provided the initial insight that complement was essential to generate an appropriate adaptive immune response. The development of knockout mice have further underlined the importance of the AP/AL in disease, with the FH knockout mouse paving the way for the first anti-complement drugs. The impact from the development of FB, properdin, and C3 knockout mice closely follows this in terms of mechanistic understanding in disease. Indeed, our current understanding that complement plays a role in most conditions at one level or another is rooted in many of these in vivo studies. That C3, in particular, has roles beyond the obvious in innate and adaptive immunity, normal physiology, and cellular functions, with or without other recognized AP components, we would argue, only extends the reach of this arm of the complement system. Humanized mouse models also continue to play their part. Here, we argue that the animal models developed over the last few decades have truly helped define the role of the AP/AL in disease.
Collapse
Affiliation(s)
- Beth G. Gibson
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| | - Thomas E. Cox
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| | - Kevin J. Marchbank
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| |
Collapse
|
31
|
Zhang XJ, Zhong YQ, Ma ZY, Hu YZ, Su JG, Zhang YA. Insights into the Antibacterial Properties of Complement Peptides C3a, C4a, and C5a across Vertebrates. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2330-2340. [PMID: 36280254 DOI: 10.4049/jimmunol.2101019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 09/30/2022] [Indexed: 02/17/2024]
Abstract
Complement peptides C3a, C4a, and C5a are important components of innate immunity in vertebrates. Although they diverged from a common ancestor, only C3a and C4a can act as antibacterial peptides in Homo sapiens, suggesting that C5a has evolved into a purely chemotactic molecule; however, the antibacterial properties of C3a, C4a, and C5a across vertebrates still require elucidation. In this article, we show that, unlike those in H. sapiens, Mus musculus C3a, C4a, and C5a all possess antibacterial activities, implying that the antibacterial properties of C3a, C4a, and C5a have evolved divergently in vertebrates. The extremely different net charge, a key factor determining the antibacterial activities of cationic antimicrobial peptides, of vertebrate C3a, C4a, and C5a supports this speculation. Moreover, the antibacterial activity of overlapping peptides covering vertebrate C3a, C4a, and C5a further strongly supports the speculation, because their activity is positively correlated with the net charge of source molecules. Notably, the structures of C3a, C4a, and C5a are conserved in vertebrates, and the inactive overlapping peptides can become antibacterial peptides if mutated to possess enough net positive charges, indicating that net charge is the only factor determining the antibacterial properties of vertebrate C3a, C4a, and C5a. More importantly, many vertebrate C3a-, C4a-, and C5a-derived peptides possess high antibacterial activities yet exhibit no hemolytic activities, suggesting the application potential in anti-infective therapy. Taken together, our findings reveal that vertebrate C3a, C4a, and C5a are all sources of antibacterial peptides that will facilitate the design of excellent peptide antibiotics.
Collapse
Affiliation(s)
- Xu-Jie Zhang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, China; and
| | - Ya-Qin Zhong
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Zi-You Ma
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Ya-Zhen Hu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Jian-Guo Su
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yong-An Zhang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, College of Fisheries, Huazhong Agricultural University, Wuhan, China;
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
32
|
Sialic acids on B cells are crucial for their survival and provide protection against apoptosis. Proc Natl Acad Sci U S A 2022; 119:e2201129119. [PMID: 35696562 PMCID: PMC9231502 DOI: 10.1073/pnas.2201129119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Sialic acids (Sias) on the B cell membrane are involved in cell migration, in the control of the complement system and, as sialic acid-binding immunoglobulin-like lectin (Siglec) ligands, in the regulation of cellular signaling. We studied the role of sialoglycans on B cells in a mouse model with B cell-specific deletion of cytidine monophosphate sialic acid synthase (CMAS), the enzyme essential for the synthesis of sialoglycans. Surprisingly, these mice showed a severe B cell deficiency in secondary lymphoid organs. Additional depletion of the complement factor C3 rescued the phenotype only marginally, demonstrating a complement-independent mechanism. The B cell survival receptor BAFF receptor was not up-regulated, and levels of activated caspase 3 and processed caspase 8 were high in B cells of Cmas-deficient mice, indicating ongoing apoptosis. Overexpressed Bcl-2 could not rescue this phenotype, pointing to extrinsic apoptosis. These results show that sialoglycans on the B cell surface are crucial for B cell survival by counteracting several death-inducing pathways.
Collapse
|
33
|
A New Role of Acute Phase Proteins: Local Production Is an Ancient, General Stress-Response System of Mammalian Cells. Int J Mol Sci 2022; 23:ijms23062972. [PMID: 35328392 PMCID: PMC8954921 DOI: 10.3390/ijms23062972] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/20/2022] [Accepted: 03/03/2022] [Indexed: 02/06/2023] Open
Abstract
The prevailing general view of acute-phase proteins (APPs) is that they are produced by the liver in response to the stress of the body as part of a systemic acute-phase response. We demonstrated a coordinated, local production of these proteins upon cell stress by the stressed cells. The local, stress-induced APP production has been demonstrated in different tissues (kidney, breast cancer) and with different stressors (hypoxia, fibrosis and electromagnetic heat). Thus, this local acute-phase response (APR) seems to be a universal mechanism. APP production is an ancient defense mechanism observed in nematodes and fruit flies as well. Local APP production at the tissue level is also supported by sporadic literature data for single proteins; however, the complex, coordinated, local appearance of this stress response has been first demonstrated only recently. Although a number of literature data are available for the local production of single acute-phase proteins, their interpretation as a local, coordinated stress response is new. A better understanding of the role of APPs in cellular stress response may also be of diagnostic/prognostic and therapeutic significance.
Collapse
|
34
|
Complement C3-targeted therapy in C3 glomerulopathy, a prototype of complement-mediated kidney diseases. Semin Immunol 2022; 60:101634. [PMID: 35817659 DOI: 10.1016/j.smim.2022.101634] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 01/15/2023]
Abstract
C3 glomerulopathy (C3G) is a rare and complex kidney disease that primarily affects young adults. Renal outcomes remain poor in the absence of specific treatment. C3G is driven by uncontrolled overactivation of the alternative complement pathway, which is mainly of acquired origin. Functional characterization of complement abnormalities (i.e., autoantibodies targeting complement components and variants in complement genes) identified in patients and experimental models of the disease improved the understanding of the disease, making C3G a prototype of complement-mediated diseases. The contribution of C3 convertase, as well as C5 convertase, in disease occurrence, phenotype, and severity is now well established, offering various potential therapeutic interventions. However, the lack of sufficient efficiency in anti-C5 therapy highlights the extreme complexity of the disease and the need for new therapeutic approaches based on C3 and C3 convertase axis inhibition. Here, we provide an overview of the complement activation mechanism involved in C3G and discuss therapeutic options based on complement inhibitors, with a specific focus on C3 inhibition.
Collapse
|
35
|
Sahin M, Remy MM, Fallet B, Sommerstein R, Florova M, Langner A, Klausz K, Straub T, Kreutzfeldt M, Wagner I, Schmidt CT, Malinge P, Magistrelli G, Izui S, Pircher H, Verbeek JS, Merkler D, Peipp M, Pinschewer DD. Antibody bivalency improves antiviral efficacy by inhibiting virion release independently of Fc gamma receptors. Cell Rep 2022; 38:110303. [PMID: 35108544 PMCID: PMC8822495 DOI: 10.1016/j.celrep.2022.110303] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/08/2021] [Accepted: 01/04/2022] [Indexed: 12/17/2022] Open
Abstract
Across the animal kingdom, multivalency discriminates antibodies from all other immunoglobulin superfamily members. The evolutionary forces conserving multivalency above other structural hallmarks of antibodies remain, however, incompletely defined. Here, we engineer monovalent either Fc-competent or -deficient antibody formats to investigate mechanisms of protection of neutralizing antibodies (nAbs) and non-neutralizing antibodies (nnAbs) in virus-infected mice. Antibody bivalency enables the tethering of virions to the infected cell surface, inhibits the release of virions in cell culture, and suppresses viral loads in vivo independently of Fc gamma receptor (FcγR) interactions. In return, monovalent antibody formats either do not inhibit virion release and fail to protect in vivo or their protective efficacy is largely FcγR dependent. Protection in mice correlates with virus-release-inhibiting activity of nAb and nnAb rather than with their neutralizing capacity. These observations provide mechanistic insights into the evolutionary conservation of antibody bivalency and help refining correlates of nnAb protection for vaccine development.
Collapse
Affiliation(s)
- Mehmet Sahin
- Department of Biomedicine - Haus Petersplatz, Division of Experimental Virology, University of Basel, 4009 Basel, Switzerland
| | - Melissa M Remy
- Department of Biomedicine - Haus Petersplatz, Division of Experimental Virology, University of Basel, 4009 Basel, Switzerland; Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Benedict Fallet
- Department of Biomedicine - Haus Petersplatz, Division of Experimental Virology, University of Basel, 4009 Basel, Switzerland; Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Rami Sommerstein
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Marianna Florova
- Department of Biomedicine - Haus Petersplatz, Division of Experimental Virology, University of Basel, 4009 Basel, Switzerland
| | - Anna Langner
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, University Hospital Schleswig-Holstein and Christian-Albrechts-University Kiel, Kiel, Germany
| | - Katja Klausz
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, University Hospital Schleswig-Holstein and Christian-Albrechts-University Kiel, Kiel, Germany
| | - Tobias Straub
- Institute for Immunology, Department for Medical Microbiology and Hygiene, University Medical Center Freiburg, 79104 Freiburg, Germany
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, Division of Clinical Pathology, University and University Hospital of Geneva, 1211 Geneva, Switzerland
| | - Ingrid Wagner
- Department of Pathology and Immunology, Division of Clinical Pathology, University and University Hospital of Geneva, 1211 Geneva, Switzerland
| | - Cinzia T Schmidt
- BioEM Lab, Center for Cellular Imaging & Nano Analytics, Biozentrum, University of Basel, Basel, Switzerland
| | - Pauline Malinge
- Light Chain Bioscience, Novimmune SA, Plan-les-Ouates, Switzerland
| | | | - Shozo Izui
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Hanspeter Pircher
- Institute for Immunology, Department for Medical Microbiology and Hygiene, University Medical Center Freiburg, 79104 Freiburg, Germany
| | - J Sjef Verbeek
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands; Department of Biomedical Engineering, Toin University of Yokohama, Yokohama, Japan
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, University and University Hospital of Geneva, 1211 Geneva, Switzerland
| | - Matthias Peipp
- Division of Stem Cell Transplantation and Immunotherapy, Department of Medicine II, University Hospital Schleswig-Holstein and Christian-Albrechts-University Kiel, Kiel, Germany
| | - Daniel D Pinschewer
- Department of Biomedicine - Haus Petersplatz, Division of Experimental Virology, University of Basel, 4009 Basel, Switzerland; Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
36
|
Lazzaro A, De Girolamo G, Filippi V, Innocenti GP, Santinelli L, Ceccarelli G, Trecarichi EM, Torti C, Mastroianni CM, d’Ettorre G, Russo A. The Interplay between Host Defense, Infection, and Clinical Status in Septic Patients: A Narrative Review. Int J Mol Sci 2022; 23:ijms23020803. [PMID: 35054993 PMCID: PMC8776148 DOI: 10.3390/ijms23020803] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/05/2022] [Accepted: 01/09/2022] [Indexed: 01/08/2023] Open
Abstract
Sepsis is a life-threatening condition that arises when the body's response to an infection injures its own tissues and organs. Despite significant morbidity and mortality throughout the world, its pathogenesis and mechanisms are not clearly understood. In this narrative review, we aimed to summarize the recent developments in our understanding of the hallmarks of sepsis pathogenesis (immune and adaptive immune response, the complement system, the endothelial disfunction, and autophagy) and highlight novel laboratory diagnostic approaches. Clinical management is also discussed with pivotal consideration for antimicrobic therapy management in particular settings, such as intensive care unit, altered renal function, obesity, and burn patients.
Collapse
Affiliation(s)
- Alessandro Lazzaro
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00161 Rome, Italy; (A.L.); (G.D.G.); (V.F.); (G.P.I.); (L.S.); (G.C.); (C.M.M.); (G.d.)
| | - Gabriella De Girolamo
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00161 Rome, Italy; (A.L.); (G.D.G.); (V.F.); (G.P.I.); (L.S.); (G.C.); (C.M.M.); (G.d.)
| | - Valeria Filippi
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00161 Rome, Italy; (A.L.); (G.D.G.); (V.F.); (G.P.I.); (L.S.); (G.C.); (C.M.M.); (G.d.)
| | - Giuseppe Pietro Innocenti
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00161 Rome, Italy; (A.L.); (G.D.G.); (V.F.); (G.P.I.); (L.S.); (G.C.); (C.M.M.); (G.d.)
| | - Letizia Santinelli
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00161 Rome, Italy; (A.L.); (G.D.G.); (V.F.); (G.P.I.); (L.S.); (G.C.); (C.M.M.); (G.d.)
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00161 Rome, Italy; (A.L.); (G.D.G.); (V.F.); (G.P.I.); (L.S.); (G.C.); (C.M.M.); (G.d.)
| | - Enrico Maria Trecarichi
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (E.M.T.); (C.T.)
| | - Carlo Torti
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (E.M.T.); (C.T.)
| | - Claudio Maria Mastroianni
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00161 Rome, Italy; (A.L.); (G.D.G.); (V.F.); (G.P.I.); (L.S.); (G.C.); (C.M.M.); (G.d.)
| | - Gabriella d’Ettorre
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00161 Rome, Italy; (A.L.); (G.D.G.); (V.F.); (G.P.I.); (L.S.); (G.C.); (C.M.M.); (G.d.)
| | - Alessandro Russo
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (E.M.T.); (C.T.)
- Correspondence:
| |
Collapse
|
37
|
Read BJ, Won L, Kraft JC, Sappington I, Aung A, Wu S, Bals J, Chen C, Lee KK, Lingwood D, King NP, Irvine DJ. Mannose-binding lectin and complement mediate follicular localization and enhanced immunogenicity of diverse protein nanoparticle immunogens. Cell Rep 2022; 38:110217. [PMID: 35021101 PMCID: PMC8805147 DOI: 10.1016/j.celrep.2021.110217] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/03/2021] [Accepted: 12/14/2021] [Indexed: 01/06/2023] Open
Abstract
Nanoparticle (NP) vaccine formulations promote immune responses through multiple mechanisms. We recently reported that mannose-binding lectin (MBL) triggers trafficking of glycosylated HIV Env-immunogen NPs to lymph node follicles. Here, we investigate effects of MBL and complement on NP forms of HIV and other viral antigens. MBL recognition of oligomannose on gp120 nanoparticles significantly increases antigen accumulation in lymph nodes and antigen-specific germinal center (GC) responses. MBL and complement also mediate follicular trafficking and enhance GC responses to influenza, HBV, and HPV particulate antigens. Using model protein nanoparticles bearing titrated levels of glycosylation, we determine that mannose patches at a minimal density of 2.1 × 10-3 mannose patches/nm2 are required to trigger follicular targeting, which increases with increasing glycan density up to at least ∼8.2 × 10-3 patches/nm2. Thus, innate immune recognition of glycans has a significant impact on humoral immunity, and these findings provide a framework for engineering glycan recognition to optimize vaccine efficacy.
Collapse
Affiliation(s)
- Benjamin J Read
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lori Won
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - John C Kraft
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Isaac Sappington
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Aereas Aung
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Shengwei Wu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Julia Bals
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Harvard University, Cambridge, MA 02139, USA
| | - Chengbo Chen
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA; Biological Physics Structure and Design Program, University of Washington, Seattle, WA 98195, USA
| | - Kelly K Lee
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, USA; Biological Physics Structure and Design Program, University of Washington, Seattle, WA 98195, USA
| | - Daniel Lingwood
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Harvard University, Cambridge, MA 02139, USA
| | - Neil P King
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Harvard University, Cambridge, MA 02139, USA; Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
38
|
Complement System in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms222413647. [PMID: 34948444 PMCID: PMC8705098 DOI: 10.3390/ijms222413647] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/10/2021] [Accepted: 12/15/2021] [Indexed: 01/02/2023] Open
Abstract
Alzheimer’s disease is a type of dementia characterized by problems with short-term memory, cognition, and difficulties with activities of daily living. It is a progressive, neurodegenerative disorder. The complement system is an ancient part of the innate immune system and comprises of more than thirty serum and membrane-bound proteins. This system has three different activating pathways and culminates into the formation of a membrane attack complex that ultimately causes target cell lysis (usually pathogens) The complement system is involved in several important functions in the central nervous system (CNS) that include neurogenesis, synaptic pruning, apoptosis, and neuronal plasticity. Here, we discuss how the complement system is involved in the effective functioning of CNS, while also contributing to chronic neuroinflammation leading to neurodegenerative disorders such as Alzheimer’s disease. We also discuss potential targets in the complement system for stopping its harmful effects via neuroinflammation and provide perspective for the direction of future research in this field.
Collapse
|
39
|
Complement component 3 from astrocytes mediates retinal ganglion cell loss during neuroinflammation. Acta Neuropathol 2021; 142:899-915. [PMID: 34487221 DOI: 10.1007/s00401-021-02366-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/13/2021] [Accepted: 08/27/2021] [Indexed: 12/14/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) characterized by varying degrees of secondary neurodegeneration. Retinal ganglion cells (RGC) are lost in MS in association with optic neuritis but the mechanisms of neuronal injury remain unclear. Complement component C3 has been implicated in retinal and cerebral synaptic pathology that may precede neurodegeneration. Herein, we examined post-mortem MS retinas, and then used a mouse model, experimental autoimmune encephalomyelitis (EAE), to examine the role of C3 in the pathogenesis of RGC loss associated with optic neuritis. First, we show extensive C3 expression in astrocytes (C3+/GFAP+ cells) and significant RGC loss (RBPMS+ cells) in post-mortem retinas from people with MS compared to retinas from non-MS individuals. A patient with progressive MS with a remote history of optic neuritis showed marked reactive astrogliosis with C3 expression in the inner retina extending into deeper layers in the affected eye more than the unaffected eye. To study whether C3 mediates retinal degeneration, we utilized global C3-/- EAE mice and found that they had less RGC loss and partially preserved neurites in the retina compared with C3+/+ EAE mice. C3-/- EAE mice had fewer axonal swellings in the optic nerve, reflecting reduced axonal injury, but had no changes in demyelination or T cell infiltration into the CNS. Using a C3-tdTomato reporter mouse line, we show definitive evidence of C3 expression in astrocytes in the retina and optic nerves of EAE mice. Conditional deletion of C3 in astrocytes showed RGC protection replicating the effects seen in the global knockouts. These data implicate astrocyte C3 expression as a critical mediator of retinal neuronal pathology in EAE and MS, and are consistent with recent studies showing C3 gene variants are associated with faster rates of retinal neurodegeneration in human disease.
Collapse
|
40
|
Dahmani M, Cook JH, Zhu JC, Riley SP. Contribution of classical complement activation and IgM to the control of Rickettsia infection. Mol Microbiol 2021; 116:1476-1488. [PMID: 34725868 PMCID: PMC8955150 DOI: 10.1111/mmi.14839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 01/02/2023]
Abstract
Pathogenic Rickettsia are obligate intracellular bacteria and the etiologic agents of many life‐threatening infectious diseases. Due to the serious nature of these infections, it is imperative to both identify the responsive immune sensory pathways and understand the associated immune mechanisms that restrict Rickettsia proliferation. Previous studies have demonstrated that the mammalian complement system is both activated during Rickettsia infection and contributes to the immune response to infection. To further define this component of the mammalian anti‐Rickettsia immune response, we sought to identify the mechanism(s) of complement activation during Rickettsia infection. We have employed a series of in vitro and in vivo models of infection to investigate the role of the classical complement activation pathway during Rickettsia infection. Depletion or elimination of complement activity demonstrates that both C1q and pre‐existing IgM contribute to complement activation; thus implicating the classical complement system in Rickettsia‐mediated complement activation. Elimination of the classical complement pathway from mice increases susceptibility to R. australis infection with both increased bacterial loads in multiple tissues and decreased immune activation markers. This study highlights the role of the classical complement pathway in immunity against Rickettsia and implicates resident Rickettsia‐responsive IgM in the response to infection.
Collapse
Affiliation(s)
- Mustapha Dahmani
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA.,Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| | - Jack H Cook
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA.,Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| | - Jinyi C Zhu
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA.,Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| | - Sean P Riley
- Department of Veterinary Medicine, University of Maryland-College Park, College Park, Maryland, USA.,Virginia-Maryland College of Veterinary Medicine, College Park, Maryland, USA
| |
Collapse
|
41
|
Zheng Z, Li YN, Jia S, Zhu M, Cao L, Tao M, Jiang J, Zhan S, Chen Y, Gao PJ, Hu W, Wang Y, Shao C, Shi Y. Lung mesenchymal stromal cells influenced by Th2 cytokines mobilize neutrophils and facilitate metastasis by producing complement C3. Nat Commun 2021; 12:6202. [PMID: 34707103 PMCID: PMC8551331 DOI: 10.1038/s41467-021-26460-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 10/05/2021] [Indexed: 01/21/2023] Open
Abstract
Pre-metastatic niche formation is critical for the colonization of disseminated cancer cells in distant organs. Here we find that lung mesenchymal stromal cells (LMSCs) at pre-metastatic stage possess potent metastasis-promoting activity. RNA-seq reveals an upregulation of complement 3 (C3) in those LMSCs. C3 is found to promote neutrophil recruitment and the formation of neutrophil extracellular traps (NETs), which facilitate cancer cell metastasis to the lungs. C3 expression in LMSCs is induced and sustained by Th2 cytokines in a STAT6-dependent manner. LMSCs-driven lung metastasis is abolished in Th1-skewing Stat6-deficient mice. Blockade of IL-4 by antibody also attenuates LMSCs-driven cancer metastasis to the lungs. Consistently, metastasis is greatly enhanced in Th2-skewing T-bet-deficient mice or in nude mice adoptively transferred with T-bet-deficient T cells. Increased C3 levels are also detected in breast cancer patients. Our results suggest that targeting the Th2-STAT6-C3-NETs cascade may reduce breast cancer metastasis to the lungs. The formation of the pre-metastatic niche enables the colonisation of disseminated cancer cells in distant organs. Here, the authors show that Th2 cytokines induce Complement 3 production in lung mesenchymal stromal cells, which recruits neutrophils and promotes the formation neutrophil extracellular traps, facilitating breast cancer cell metastasis to the lungs.
Collapse
Affiliation(s)
- Zhiyuan Zheng
- The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine of Soochow University, Suzhou, Jiangsu, China.,Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Cancer Center, Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ya-Nan Li
- The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine of Soochow University, Suzhou, Jiangsu, China
| | - Shanfen Jia
- The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine of Soochow University, Suzhou, Jiangsu, China
| | - Mengting Zhu
- The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine of Soochow University, Suzhou, Jiangsu, China
| | - Lijuan Cao
- The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine of Soochow University, Suzhou, Jiangsu, China
| | - Min Tao
- The First Affiliated Hospital of Soochow University/The First People's Hospital of Suzhou, Suzhou, Jiangsu, China
| | - Jingting Jiang
- The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine of Soochow University, Suzhou, Jiangsu, China
| | - Shenghua Zhan
- The First Affiliated Hospital of Soochow University/The First People's Hospital of Suzhou, Suzhou, Jiangsu, China
| | - Yongjing Chen
- The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine of Soochow University, Suzhou, Jiangsu, China
| | - Ping-Jin Gao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Weiguo Hu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Collaborative Innovation Center of Cancer Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Changshun Shao
- The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine of Soochow University, Suzhou, Jiangsu, China.
| | - Yufang Shi
- The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine of Soochow University, Suzhou, Jiangsu, China. .,The First Affiliated Hospital of Soochow University/The First People's Hospital of Suzhou, Suzhou, Jiangsu, China. .,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
42
|
Peterson SL, Li Y, Sun CJ, Wong KA, Leung KS, de Lima S, Hanovice NJ, Yuki K, Stevens B, Benowitz LI. Retinal Ganglion Cell Axon Regeneration Requires Complement and Myeloid Cell Activity within the Optic Nerve. J Neurosci 2021; 41:8508-8531. [PMID: 34417332 PMCID: PMC8513703 DOI: 10.1523/jneurosci.0555-21.2021] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/21/2021] [Accepted: 08/16/2021] [Indexed: 01/01/2023] Open
Abstract
Axon regenerative failure in the mature CNS contributes to functional deficits following many traumatic injuries, ischemic injuries, and neurodegenerative diseases. The complement cascade of the innate immune system responds to pathogen threat through inflammatory cell activation, pathogen opsonization, and pathogen lysis, and complement is also involved in CNS development, neuroplasticity, injury, and disease. Here, we investigated the involvement of the classical complement cascade and microglia/monocytes in CNS repair using the mouse optic nerve injury (ONI) model, in which axons arising from retinal ganglion cells (RGCs) are disrupted. We report that central complement C3 protein and mRNA, classical complement C1q protein and mRNA, and microglia/monocyte phagocytic complement receptor CR3 all increase in response to ONI, especially within the optic nerve itself. Importantly, genetic deletion of C1q, C3, or CR3 attenuates RGC axon regeneration induced by several distinct methods, with minimal effects on RGC survival. Local injections of C1q function-blocking antibody revealed that complement acts primarily within the optic nerve, not retina, to support regeneration. Moreover, C1q opsonizes and CR3+ microglia/monocytes phagocytose growth-inhibitory myelin debris after ONI, a likely mechanism through which complement and myeloid cells support axon regeneration. Collectively, these results indicate that local optic nerve complement-myeloid phagocytic signaling is required for CNS axon regrowth, emphasizing the axonal compartment and highlighting a beneficial neuroimmune role for complement and microglia/monocytes in CNS repair.SIGNIFICANCE STATEMENT Despite the importance of achieving axon regeneration after CNS injury and the inevitability of inflammation after such injury, the contributions of complement and microglia to CNS axon regeneration are largely unknown. Whereas inflammation is commonly thought to exacerbate the effects of CNS injury, we find that complement proteins C1q and C3 and microglia/monocyte phagocytic complement receptor CR3 are each required for retinal ganglion cell axon regeneration through the injured mouse optic nerve. Also, whereas studies of optic nerve regeneration generally focus on the retina, we show that the regeneration-relevant role of complement and microglia/monocytes likely involves myelin phagocytosis within the optic nerve. Thus, our results point to the importance of the innate immune response for CNS repair.
Collapse
Affiliation(s)
- Sheri L Peterson
- Laboratories for Neuroscience Research in Neurosurgery
- Department of Neurosurgery
- F.M. Kirby Neurobiology Center, and
- Department of Neurosurgery and
| | - Yiqing Li
- Laboratories for Neuroscience Research in Neurosurgery
- Department of Neurosurgery
- F.M. Kirby Neurobiology Center, and
- Department of Neurosurgery and
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong China, 510060
| | - Christina J Sun
- Laboratories for Neuroscience Research in Neurosurgery
- Department of Neurosurgery
| | - Kimberly A Wong
- Laboratories for Neuroscience Research in Neurosurgery
- Department of Neurosurgery
- F.M. Kirby Neurobiology Center, and
- Department of Neurosurgery and
| | - Kylie S Leung
- Laboratories for Neuroscience Research in Neurosurgery
- Department of Neurosurgery
| | - Silmara de Lima
- Laboratories for Neuroscience Research in Neurosurgery
- Department of Neurosurgery
- F.M. Kirby Neurobiology Center, and
- Department of Neurosurgery and
| | - Nicholas J Hanovice
- Laboratories for Neuroscience Research in Neurosurgery
- Department of Neurosurgery
- F.M. Kirby Neurobiology Center, and
- Department of Neurosurgery and
| | - Kenya Yuki
- Laboratories for Neuroscience Research in Neurosurgery
- Department of Neurosurgery
- F.M. Kirby Neurobiology Center, and
- Department of Neurosurgery and
| | - Beth Stevens
- F.M. Kirby Neurobiology Center, and
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts 02115
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142
| | - Larry I Benowitz
- Laboratories for Neuroscience Research in Neurosurgery
- Department of Neurosurgery
- F.M. Kirby Neurobiology Center, and
- Department of Neurosurgery and
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
43
|
DeKorver NW, Chaudoin TR, Zhao G, Wang D, Arikkath J, Bonasera SJ. Complement Component C3 Loss leads to Locomotor Deficits and Altered Cerebellar Internal Granule Cell In Vitro Synaptic Protein Expression in C57BL/6 Mice. Mol Neurobiol 2021; 58:5857-5875. [PMID: 34415487 DOI: 10.1007/s12035-021-02480-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/24/2021] [Indexed: 01/14/2023]
Abstract
Complement component 3 (C3) expression is increased in the cerebellum of aging mice that demonstrate locomotor impairments and increased excitatory synapse density. However, C3 regulation of locomotion, as well as C3 roles in excitatory synapse function, remains poorly understood. Here, we demonstrate that constitutive loss of C3 function in mice evokes a locomotor phenotype characterized by decreased speed, increased active state locomotor probability, and gait ataxia. C3 loss does not alter metabolism or body mass composition. No evidence of significant muscle weakness or degenerative arthritis was found in C3 knockout mice to explain decreased gait speeds. In an enriched primary cerebellar granule cell culture model, loss of C3 protein results in increased excitatory synaptic density and increased response to KCl depolarization. Our analysis of excitatory synaptic density in the cerebellar internal granule cell and molecular layers did not demonstrate increased synaptic density in vivo, suggesting the presence of compensatory mechanisms regulating synaptic development. Functional deficits in C3 knockout mice are therefore more likely to result from altered synaptic function and/or connectivity than gross synaptic deficits. Our data demonstrate a novel role for complement proteins in cerebellar regulation of locomotor output and control.
Collapse
Affiliation(s)
- Nicholas W DeKorver
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, 3028 Durham Research Center II, Omaha, NE, 68198-5039, USA
| | - Tammy R Chaudoin
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, 3028 Durham Research Center II, Omaha, NE, 68198-5039, USA
| | - Gang Zhao
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 986125 Nebraska Medical Center, PDD 3020, Omaha, NE, 68198-6125, USA
| | - Dong Wang
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, 986125 Nebraska Medical Center, PDD 3020, Omaha, NE, 68198-6125, USA
| | - Jyothi Arikkath
- Monroe-Meyer Institute, University of Nebraska Medical Center, 3031 Durham Research Center II, Omaha, NE, 68198-5960, USA
| | - Stephen J Bonasera
- Department of Internal Medicine, Division of Geriatrics, University of Nebraska Medical Center, 3028 Durham Research Center II, Omaha, NE, 68198-5039, USA.
| |
Collapse
|
44
|
Agostinis C, Zorzet S, Balduit A, Zito G, Mangogna A, Macor P, Romano F, Toffoli M, Belmonte B, Morello G, Martorana A, Borelli V, Ricci G, Kishore U, Bulla R. The Inflammatory Feed-Forward Loop Triggered by the Complement Component C3 as a Potential Target in Endometriosis. Front Immunol 2021; 12:693118. [PMID: 34489939 PMCID: PMC8418148 DOI: 10.3389/fimmu.2021.693118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/26/2021] [Indexed: 12/25/2022] Open
Abstract
The complement system is a major component of humoral innate immunity, acting as a first line of defense against microbes via opsonization and lysis of pathogens. However, novel roles of the complement system in inflammatory and immunological processes, including in cancer, are emerging. Endometriosis (EM), a benign disease characterized by ectopic endometrial implants, shows certain unique features of cancer, such as the capacity to invade surrounding tissues, and in severe cases, metastatic properties. A defective immune surveillance against autologous tissue deposited in the peritoneal cavity allows immune escape for endometriotic lesions. There is evidence that the glandular epithelial cells found in endometriotic implants produce and secrete the complement component C3. Here, we show, using immunofluorescence and RT-qPCR, the presence of locally synthesized C3 in the ectopic endometriotic tissue, but not in the eutopic tissue. We generated a murine model of EM via injection of minced uterine tissue from a donor mouse into the peritoneum of recipient mice. The wild type mice showed greater amount of cyst formation in the peritoneum compared to C3 knock-out mice. Peritoneal washings from the wild type mice with EM showed more degranulated mast cells compared to C3 knock-out mice, consistent with higher C3a levels in the peritoneal fluid of EM patients. We provide evidence that C3a participates in an auto-amplifying loop leading to mast cell infiltration and activation, which is pathogenic in EM. Thus, C3 can be considered a marker of EM and its local synthesis can promote the engraftment of the endometriotic cysts.
Collapse
Affiliation(s)
- Chiara Agostinis
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Sonia Zorzet
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Andrea Balduit
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Gabriella Zito
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Alessandro Mangogna
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Federico Romano
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Miriam Toffoli
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Human Pathology Section, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Gaia Morello
- Tumor Immunology Unit, Human Pathology Section, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Anna Martorana
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Violetta Borelli
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Giuseppe Ricci
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
- Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
45
|
Bikhet M, Iwase H, Yamamoto T, Jagdale A, Foote JB, Ezzelarab M, Anderson DJ, Locke JE, Eckhoff DE, Hara H, Cooper DKC. What Therapeutic Regimen Will Be Optimal for Initial Clinical Trials of Pig Organ Transplantation? Transplantation 2021; 105:1143-1155. [PMID: 33534529 DOI: 10.1097/tp.0000000000003622] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We discuss what therapeutic regimen might be acceptable/successful in the first clinical trial of genetically engineered pig kidney or heart transplantation. As regimens based on a calcineurin inhibitor or CTLA4-Ig have proved unsuccessful, the regimen we administer to baboons is based on induction therapy with antithymocyte globulin, an anti-CD20 mAb (Rituximab), and cobra venom factor, with maintenance therapy based on blockade of the CD40/CD154 costimulation pathway (with an anti-CD40 mAb), with rapamycin, and a corticosteroid. An anti-inflammatory agent (etanercept) is administered for the first 2 wk, and adjuvant therapy includes prophylaxis against thrombotic complications, anemia, cytomegalovirus, and pneumocystis. Using this regimen, although antibody-mediated rejection certainly can occur, we have documented no definite evidence of an adaptive immune response to the pig xenograft. This regimen could also form the basis for the first clinical trial, except that cobra venom factor will be replaced by a clinically approved agent, for example, a C1-esterase inhibitor. However, none of the agents that block the CD40/CD154 pathway are yet approved for clinical use, and so this hurdle remains to be overcome. The role of anti-inflammatory agents remains unproven. The major difference between this suggested regimen and those used in allotransplantation is the replacement of a calcineurin inhibitor with a costimulation blockade agent, but this does not appear to increase the complications of the regimen.
Collapse
Affiliation(s)
- Mohamed Bikhet
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Hayato Iwase
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Takayuki Yamamoto
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Abhijit Jagdale
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Jeremy B Foote
- Department of Microbiology and Animal Resources Program, University of Alabama at Birmingham, Birmingham, AL
| | - Mohamed Ezzelarab
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Douglas J Anderson
- Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Jayme E Locke
- Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Devin E Eckhoff
- Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - Hidetaka Hara
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| | - David K C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
46
|
Abstract
BACKGROUND Circulating complement C3 fragments released during septic shock might contribute to the development of complications such as profound hypotension and disseminated intravascular coagulation. The role of C3 in the course of septic shock varies in the literature, possibly because circulating C3 exists in different forms indistinguishable via traditional ELISA-based methods. We sought to test the relationship between C3 forms, measured by Western blotting with its associated protein size differentiation feature, and clinical outcomes. METHODS Secondary analysis of two prospective cohorts of patients with septic shock: a discovery cohort of 24 patents and a validation cohort of 181 patients. C3 levels were measured by Western blotting in both cohorts using blood obtained at enrollment. Differences between survivors and non-survivors were compared, and the independent prognostic values of C3 forms were assessed. RESULTS In both cohorts there were significantly lower levels of the C3-alpha chain in non-survivors than in survivors, and persisted after controlling for sequential organ failure assessment score. Area under the receiver operating characteristics to predict survival was 0.65 (95% confidence interval: 0.56-0.75). At a best cutoff value (Youden) of 970.6 μg/mL, the test demonstrated a sensitivity of 68.5% and specificity of 61.5%. At this cutoff point, Kaplan-Meier survival analysis showed that patients with lower levels of C3-alpha chain had significantly lower survival than those with higher levels (P < 0.001). CONCLUSION Circulating C3-alpha chain levels is a significant independent predictor of survival in septic shock patients.
Collapse
|
47
|
Moon CJ, Kwon TH, Lee KS, Lee HS. Recurrent neonatal sepsis and progressive white matter injury in a premature newborn culture-positive for group B Streptococcus: A case report. Medicine (Baltimore) 2021; 100:e26387. [PMID: 34160417 PMCID: PMC8238304 DOI: 10.1097/md.0000000000026387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/02/2021] [Indexed: 01/04/2023] Open
Abstract
RATIONALE Group B Streptococcus (GBS) remains a principal pathogen causing neonatal sepsis and meningitis, particularly in premature infants with relatively insufficient immunity. Recurrence may occur uncommonly, largely associated with subclinical mucosal persistence or repetitive exposure to exogenous sources. White matter injury (WMI) including cystic periventricular leukomalacia (PVL) has been associated with intrauterine infection/inflammation, and neonatal infection as a more significant predictor including postnatal sepsis and recurrent infection, even without microbial neuroinvasion. Furthermore, clinical and experimental evidence of WMI by some bacteria other than GBS without central nervous system invasion has been reported. However, there is little evidence of WMI associated with neonatal GBS sepsis in the absence of meningitis in the literature. PATIENT CONCERNS A newborn at 30+4 weeks' gestation with low birthweight presented with 2 episodes (with a 13-day interval with no antibiotic therapy) of neonatal sepsis culture-proven for GBS with early-onset presentation after clinical chorioamnionitis via vertical GBS transmission and the associated conditions including prematurity-related neonatal immunodeficiency and persistent mucosal GBS carriage after the first antibiotic treatment. The perinatal GBS infection was complicated by progressive WMI presenting with ventriculomegaly and cystic PVL without a definite evidence of meningitis, intraventricular hemorrhage, and documented cerebral hypoxia or hypoperfusion conditions including septic shock. DIAGNOSES Recurrent group B streptococcal sepsis and cystic PVL with ventriculomegaly. INTERVENTIONS Two episodes of GBS sepsis were treated with 15-day parenteral antibiotic therapy, respectively. OUTCOMES Resolution of the recurrent GBS sepsis without further relapses, however, complicated by WMI and subsequent about 6 months delay in motor development at 12 months' corrected age. LESSONS This case suggests WMI associated with GBS bacteremia without central nervous system entry by viable GBS and also shows that in premature infants, intrauterine GBS infection with no interventions may lead to extensive and persistent GBS colonization, early-onset and recurrent GBS disease, and WMI. Postnatal as well as intrauterine infection/inflammation controls with maternal prophylaxis may be pivotal for prevention and limiting the magnitude of neurologic injury.
Collapse
MESH Headings
- Administration, Intravenous
- Anti-Bacterial Agents/administration & dosage
- Chorioamnionitis/diagnosis
- Chorioamnionitis/microbiology
- Developmental Disabilities/diagnosis
- Developmental Disabilities/microbiology
- Drug Therapy, Combination/methods
- Female
- Humans
- Hydrocephalus/diagnosis
- Hydrocephalus/microbiology
- Infant
- Infant, Low Birth Weight
- Infant, Newborn
- Infant, Premature
- Infectious Disease Transmission, Vertical
- Leukomalacia, Periventricular/diagnosis
- Leukomalacia, Periventricular/microbiology
- Leukomalacia, Periventricular/pathology
- Magnetic Resonance Imaging
- Male
- Maternal Age
- Neonatal Sepsis/diagnosis
- Neonatal Sepsis/microbiology
- Neonatal Sepsis/therapy
- Pregnancy
- Pregnancy Complications, Infectious/diagnosis
- Pregnancy Complications, Infectious/microbiology
- Recurrence
- Streptococcal Infections/complications
- Streptococcal Infections/diagnosis
- Streptococcal Infections/microbiology
- Streptococcal Infections/transmission
- Streptococcus agalactiae/isolation & purification
- White Matter/diagnostic imaging
- White Matter/microbiology
- White Matter/pathology
- Young Adult
Collapse
Affiliation(s)
- Cheong-Jun Moon
- Department of Pediatrics, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea
| | - Tae Hee Kwon
- Department of Radiology, Cha Gangnam Medical Center, Cha University School of Medicine
| | - Kyung Sang Lee
- Department of Radiology, Cha Gangnam Medical Center, Cha University School of Medicine
| | - Hyun-Seung Lee
- Department of Pediatrics, Incheon Worker‘s Compensation Hospital, Incheon
- Department of Pediatrics, Cha Gangnam Medical Center, Cha University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
48
|
Byrne RAJ, Torvell M, Daskoulidou N, Fathalla D, Kokkali E, Carpanini SM, Morgan BP. Novel Monoclonal Antibodies Against Mouse C1q: Characterisation and Development of a Quantitative ELISA for Mouse C1q. Mol Neurobiol 2021; 58:4323-4336. [PMID: 34002346 PMCID: PMC8487419 DOI: 10.1007/s12035-021-02419-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/30/2021] [Indexed: 12/20/2022]
Abstract
Recent studies have identified roles for complement in synaptic pruning, both physiological during development and pathological in Alzheimer's disease (AD). These reports suggest that C1q initiates complement activation on synapses and C3 fragments then tag them for removal by microglia. There is an urgent need to characterise these processes in rodent AD models; this requires the development of reagents and methods for detection and quantification of rodent C1q in fluids and pathological tissues. These will enable better evaluation of the role of C1q in disease and its value as disease biomarker. We describe the generation in C1q-deficient mice of novel monoclonal antibodies against mouse and rat C1q that enabled development of a sensitive, specific, and quantitative ELISA for mouse and rat C1q capable of measuring C1q in biological fluids and tissue extracts. Serum C1q levels were measured in wild-type (WT), C1q knockout (KO), C3 KO, C7 KO, Crry KO, and 3xTg and APPNL-G-F AD model mice through ageing. C1q levels significantly decreased in WT, APPNL-G-F, and C7 KO mice with ageing. C1q levels were reduced in APPNL-G-F compared to WT at all ages and in 3xTg at 12 months; C3 KO and C7 KO, but not Crry KO mice, also demonstrated significantly lower C1q levels compared to matched WT. In brain homogenates, C1q levels increased with age in both WT and APPNL-G-F mice. This robust and adaptable assay for quantification of mouse and rat C1q provides a vital tool for investigating the expression of C1q in rodent models of AD and other complement-driven pathologies.
Collapse
Affiliation(s)
- Robert A J Byrne
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK.,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK
| | - Megan Torvell
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK.,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK
| | - Nikoleta Daskoulidou
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK.,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK
| | - Dina Fathalla
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK.,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK
| | - Eirini Kokkali
- School of Optometry and Visual Sciences, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK
| | - Sarah M Carpanini
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK.,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK
| | - B Paul Morgan
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK. .,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK.
| |
Collapse
|
49
|
Anisuzzaman, Frahm S, Prodjinotho UF, Bhattacharjee S, Verschoor A, Prazeres da Costa C. Host-Specific Serum Factors Control the Development and Survival of Schistosoma mansoni. Front Immunol 2021; 12:635622. [PMID: 33968028 PMCID: PMC8103320 DOI: 10.3389/fimmu.2021.635622] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 04/06/2021] [Indexed: 11/28/2022] Open
Abstract
Introduction Schistosomiasis is a neglected tropical disease (NTD) caused by blood-dwelling flatworms which develop from skin-penetrating cercariae, the freely swimming water-borne infective stage of Schistosoma mansoni, into adult worms. This natural course of infection can be mimicked in experimental mouse models of schistosomiasis. However, only a maximum of 20-30% of penetrated cercariae mature into fecund adults. The reasons for this are unknown but could potentially involve soluble factors of the innate immune system, such as complement factors and preexisting, natural antibodies. Materials and Methods Using our recently developed novel serum- and cell-free in vitro culture system for newly transformed schistosomula (NTS), which supports long-term larval survival, we investigated the effects of mouse serum and its major soluble complement factors C1q, C3, C4 as well as preexisting, natural IgM in vitro and assessed worm development in vivo by infecting complement and soluble (s)IgM-deficient animals. Results In contrast to sera from humans and a broad variety of mammalian species, serum from mice, surprisingly, killed parasites already at skin stage in vitro. Interestingly, the most efficient killing component(s) were heat-labile but did not include important members of the perhaps best known family of heat-labile serum factors, the complement system, nor consisted of complement-activating natural immunoglobulins. Infection of complement C1q and sIgM-deficient mice with S. mansoni as well as in vitro tests with sera from mice deficient in C3 and C4 revealed no major role for these soluble factors in vivo in regard to parasite maturation, fecundity and associated immunopathology. Rather, the reduction of parasite maturation from cercariae to adult worms was comparable to wild-type mice. Conclusion This study reveals that not yet identified heat-labile serum factors are major selective determinants of the host-specificity of schistosomiasis, by directly controlling schistosomal development and survival.
Collapse
Affiliation(s)
- Anisuzzaman
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
- Department of Parasitology, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Sören Frahm
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
| | - Ulrich Fabien Prodjinotho
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
| | - Sonakshi Bhattacharjee
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
| | - Admar Verschoor
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Clarissa Prazeres da Costa
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
- Centre for Global Health, Technical University of Munich (TUM), Munich, Germany
| |
Collapse
|
50
|
Chen Y, Wang Z, Yang Z, Yang Y, Yang J, Han H, Yang H. The effect of different dietary levels of sodium and chloride on performance, blood parameters and excreta quality in goslings at 29 to 70 days of age. J Anim Physiol Anim Nutr (Berl) 2021; 106:98-109. [PMID: 33844875 DOI: 10.1111/jpn.13541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 01/09/2023]
Abstract
The purpose of this study was to ascertain the appropriate levels of dietary sodium (Na+ ) and chloride (Cl- ) for 29- to 70-day-old goslings and to investigate the effects of different levels of Na+ and Cl- on the growth performance, water consumption, blood parameters and excreta quality of goslings to provide a reference for the healthy production of goslings. In Experiment 1, a total of 432 29-day-old male Jiangnan White goslings were randomly allotted to nine treatments according to a 3 × 3 factorial design, with six pens containing eight birds per treatment. The goslings were fed diets with three concentrations of Na+ (0.10%, 0.15% and 0.20%) and three concentrations of Cl- (0.15%, 0.20% and 0.25%). The experimental period was 42 days. In Experiment 2, a total of 24 70-day-old Jiangnan White goslings were selected for four treatments (0.10% Na+ × 0.15% Cl- ; 0.10% Na+ × 0.25% Cl- ; 0.20% Na+ × 0.15% Cl- and 0.20% Na+ × 0.25% Cl- ) and housed separately in metabolic cages. The faeces were collected for 3 consecutive days. In Experiment 1, the average daily feed intake (ADFI), average daily gain (ADG) and feed/gain (F/G) ratio of goslings were unaffected by the treatments. However, low levels of Na+ and Cl- significantly reduced the water consumption of goslings in the later growth period (p < 0.05). The average water consumption of goslings fed with 0.10% Na+ × 0.15% Cl- was significantly lower than that of the goslings fed with 0.20% Na+ × 0.25% Cl- (56 days, 1304.2 ml vs. 1471.7 ml; 63 days, 1452.8 ml vs. 1610.8 ml; 70 days, 1540.0 ml vs. 1775.4 ml; p < 0.05). The interaction between Na+ and Cl- (Na+ × Cl- ) had a significant impact on the blood haemoglobin (HGB) and haematocrit (HCT) levels in the goslings (p < 0.05). HGB increased linearly with increasing levels of Na+ . HGB and HCT first increased and then decreased with increasing levels of Cl- . In Experiment 2, Na+ and Cl- levels had significant effects on the excreta moisture content (p < 0.05). Goslings fed with 0.10% Na+ × 0.15% Cl- had a low moisture content of 5.58% compared to the goslings fed with 0.20% Na+ × 0.25% Cl- (87.51% vs. 93.09%; p < 0.05). The levels of dietary Na+ had a significant effect on the retention ratio of Na (p < 0.05), with the value for the 0.20% Na+ group being significantly higher than that for the 0.10% Na+ group (p < 0.05). In summary, different levels of Na+ and Cl- did not affect the growth of goslings. To reduce the water consumption and moisture content of excreta, the Na+ and Cl- levels in the diet can be as low as 0.10% and 0.15%, respectively.
Collapse
Affiliation(s)
| | | | - Zhi Yang
- Yangzhou University, Yangzhou, China
| | - Yu Yang
- Yangzhou University, Yangzhou, China
| | | | - Houming Han
- Jiangsu Lihua Animal Husbandry Co., Ltd, Changzhou, China
| | | |
Collapse
|