1
|
Smirnov A, Daily KP, Gray MC, Ragland SA, Werner LM, Brittany Johnson M, Eby JC, Hewlett EL, Taylor RP, Criss AK. Phagocytosis via complement receptor 3 enables microbes to evade killing by neutrophils. J Leukoc Biol 2023; 114:1-20. [PMID: 36882066 PMCID: PMC10949953 DOI: 10.1093/jleuko/qiad028] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
CR3 (CD11b/CD18; αmβ2 integrin) is a conserved phagocytic receptor. The active conformation of CR3 binds the iC3b fragment of complement C3 as well as many host and microbial ligands, leading to actin-dependent phagocytosis. There are conflicting reports about how CR3 engagement affects the fate of phagocytosed substrates. Using imaging flow cytometry, we confirmed that binding and internalization of iC3b-opsonized polystyrene beads by primary human neutrophils was CR3-dependent. iC3b-opsonized beads did not stimulate neutrophil reactive oxygen species, and most beads were found in primary granule-negative phagosomes. Similarly, Neisseria gonorrhoeae that does not express phase-variable Opa proteins suppresses neutrophil reactive oxygen species and delays phagolysosome formation. Here, binding and internalization of Opa-deleted (Δopa) N. gonorrhoeae by adherent human neutrophils was inhibited using blocking antibodies against CR3 and by adding neutrophil inhibitory factor, which targets the CD11b I-domain. No detectable C3 was deposited on N. gonorrhoeae in the presence of neutrophils alone. Conversely, overexpressing CD11b in HL-60 promyelocytes enhanced Δopa N. gonorrhoeae phagocytosis, which required the CD11b I-domain. Phagocytosis of N. gonorrhoeae was also inhibited in mouse neutrophils that were CD11b-deficient or treated with anti-CD11b. Phorbol ester treatment upregulated surface CR3 on neutrophils in suspension, enabling CR3-dependent phagocytosis of Δopa N. gonorrhoeae. Neutrophils exposed to Δopa N. gonorrhoeae had limited phosphorylation of Erk1/2, p38, and JNK. Neutrophil phagocytosis of unopsonized Mycobacterium smegmatis, which also resides in immature phagosomes, was CR3-dependent and did not elicit reactive oxygen species. We suggest that CR3-mediated phagocytosis is a silent mode of entry into neutrophils, which is appropriated by diverse pathogens to subvert phagocytic killing.
Collapse
Affiliation(s)
- Asya Smirnov
- Department of Microbiology, Immunology, and Cancer Biology
| | | | - Mary C. Gray
- Department of Microbiology, Immunology, and Cancer Biology
| | | | | | | | - Joshua C. Eby
- Division of Infectious Diseases and International Health, Department of Medicine
| | - Erik L. Hewlett
- Division of Infectious Diseases and International Health, Department of Medicine
| | - Ronald P. Taylor
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine
| | | |
Collapse
|
2
|
Hao H, Wu S, Lin J, Zheng Z, Zhou Y, Zhang Y, Guo Q, Tian F, Zhao M, Chen Y, Xu X, Hou L, Wang X, Tang R. Immunization against Zika by entrapping live virus in a subcutaneous self-adjuvanting hydrogel. Nat Biomed Eng 2023; 7:928-942. [PMID: 36959404 DOI: 10.1038/s41551-023-01014-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 02/20/2023] [Indexed: 03/25/2023]
Abstract
The threat of new viral outbreaks has heightened the need for ready-to-use vaccines that are safe and effective. Here we show that a subcutaneous vaccine consisting of live Zika virus electrostatically entrapped in a self-adjuvanting hydrogel recruited immune cells at the injection site and provided mice with effective protection against a lethal viral challenge. The hydrogel prevented the escape of the viral particles and upregulated pattern recognition receptors that activated innate antiviral immunity. The local inflammatory niche facilitated the engulfment of the virus by immune cells infiltrating the hydrogel, the processing and cross-presentation of antigens and the expansion of germinal centre B cells and induced robust antigen-specific adaptive responses and immune memory. Inflammatory immune niches entrapping live viruses may facilitate the rapid development of safe and efficacious vaccines.
Collapse
Affiliation(s)
- Haibin Hao
- Department of Chemistry, Zhejiang University, Hangzhou, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, Beijing, China
| | - Jiake Lin
- Department of Chemistry, Zhejiang University, Hangzhou, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Zitong Zheng
- Department of Chemistry, Zhejiang University, Hangzhou, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Yuemin Zhou
- Department of Chemistry, Zhejiang University, Hangzhou, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Ying Zhang
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Qiang Guo
- Beijing Institute of Biotechnology, Beijing, China
| | - Fengchao Tian
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Mengsu Zhao
- Beijing Institute of Biotechnology, Beijing, China
| | - Yi Chen
- Beijing Institute of Biotechnology, Beijing, China
| | - Xurong Xu
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, Beijing, China.
| | - Xiaoyu Wang
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China.
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Ruikang Tang
- Department of Chemistry, Zhejiang University, Hangzhou, China.
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
3
|
Liebold I, Meyer S, Heine M, Kuhl A, Witt J, Eissing L, Fischer AW, Koop AC, Kluwe J, zur Wiesch JS, Wehmeyer M, Knippschild U, Scheja L, Heeren J, Bosurgi L, Worthmann A. TREM2 Regulates the Removal of Apoptotic Cells and Inflammatory Processes during the Progression of NAFLD. Cells 2023; 12:cells12030341. [PMID: 36766683 PMCID: PMC9913311 DOI: 10.3390/cells12030341] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/09/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver pathology worldwide. In mice and humans, NAFLD progression is characterized by the appearance of TREM2-expressing macrophages in the liver. However, their mechanistic contributions to disease progression have not been completely elucidated. Here, we show that TREM2+ macrophages prevent the generation of a pro-inflammatory response elicited by LPS-laden lipoproteins in vitro. Further, Trem2 expression regulates bone-marrow-derived macrophages (BMDMs) and Kupffer cell capacity to phagocyte apoptotic cells in vitro, which is dependent on CD14 activation. In line with this, loss of Trem2 resulted in an increased pro-inflammatory response, which ultimately aggravated liver fibrosis in murine models of NAFLD. Similarly, in a human NAFLD cohort, plasma levels of TREM2 were increased and hepatic TREM2 expression was correlated with higher levels of liver triglycerides and the acquisition of a fibrotic gene signature. Altogether, our results suggest that TREM2+ macrophages have a protective function during the progression of NAFLD, as they are involved in the processing of pro-inflammatory lipoproteins and phagocytosis of apoptotic cells and, thereby, are critical contributors for the re-establishment of liver homeostasis.
Collapse
Affiliation(s)
- Imke Liebold
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
| | - Simon Meyer
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Anastasia Kuhl
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Jennifer Witt
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Leah Eissing
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Alexander W. Fischer
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Anja Christina Koop
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Johannes Kluwe
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Department of Internal Medicine and Gastroenterology, Amalie Sieveking Hospital, 22359 Hamburg, Germany
| | | | - Malte Wehmeyer
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, University Hospital Ulm, 89081 Ulm, Germany
| | - Ludger Scheja
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Joerg Heeren
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lidia Bosurgi
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany
- Correspondence: (L.B.); (A.W.)
| | - Anna Worthmann
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Correspondence: (L.B.); (A.W.)
| |
Collapse
|
4
|
Mechanisms of selective monocyte targeting by liposomes functionalized with a cationic, arginine-rich lipopeptide. Acta Biomater 2022; 144:96-108. [PMID: 35314364 DOI: 10.1016/j.actbio.2022.03.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 03/08/2022] [Accepted: 03/13/2022] [Indexed: 01/01/2023]
Abstract
Stimulation of monocytes with immunomodulating agents can harness the immune system to treat a long range of diseases, including cancers, infections and autoimmune diseases. To this end we aimed to develop a monocyte-targeting delivery platform based on cationic liposomes, which can be utilized to deliver immunomodulators and thus induce monocyte-mediated immune responses while avoiding off-target side-effects. The cationic liposome design is based on functionalizing the liposomal membrane with a cholesterol-anchored tri-arginine peptide (TriArg). We demonstrate that TriArg liposomes can target monocytes with high specificity in both human and murine blood and that this targeting is dependent on the content of TriArg in the liposomal membrane. In addition, we show that the mechanism of selective monocyte targeting involves the CD14 co-receptor, and selectivity is compromised when the TriArg content is increased, resulting in complement-mediated off-target uptake in granulocytes. The presented mechanistic findings of uptake by peripheral blood leukocytes may guide the design of future drug delivery systems utilized for immunotherapy. STATEMENT OF SIGNIFICANCE: Monocytes are attractive targets for immunotherapies of cancers, infections and autoimmune diseases. Specific delivery of immunostimulatory drugs to monocytes is typically achieved using ligand-targeted drug delivery systems, but a simpler approach is to target monocytes using cationic liposomes. To achieve this, however, a deep understanding of the mechanisms governing the interactions of cationic liposomes with monocytes and other leukocytes is required. We here investigate these interactions using liposomes incorporating a cationic arginine-rich lipopeptide. We demonstrate that monocyte targeting can be achieved by fine-tuning the lipopeptide content in the liposomes. Additionally, we reveal that the CD14 receptor is involved in the targeting process, whereas the complement system is not. These mechanistic findings are critical for future design of monocyte-targeting liposomal therapies.
Collapse
|
5
|
Rivolta AA, Pittman DC, Kappes AJ, Stancil RK, Kogan C, Sanz MG. The type of anticoagulant used for plasma collection affects in vitro Rhodococcus equi assays. BMC Res Notes 2022; 15:50. [PMID: 35164828 PMCID: PMC8842809 DOI: 10.1186/s13104-022-05933-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 01/28/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Objective
The efficacy of Rhodococcus equi-specific hyperimmune plasma (HIP) is usually evaluated in vitro. Anticoagulants (AC) used for plasma collection can negatively impact bacterial replication but their effect on R. equi growth has not been evaluated. The aim of this study was to establish the effect that AC routinely used in veterinary medicine (ACD, K2EDTA, Li Heparin, and Na Citrate) have on in vitro R. equi growth. To assess this, in vitro assays commonly used to test HIP efficacy (direct effect on microorganism and macrophage infection), were performed using each AC and non-treated bacteria.
Results
There was no direct effect of ACD, Li Heparin or Na Citrate on R. equi growth. These AC significantly (p < 0.05) delayed growth for 12 h following opsonization. The number of R. equi colonies after macrophage infection was significantly (p < 0.05) lower 72 h post-opsonization with Na Citrate. K2EDTA inhibited the formation of R. equi colonies by 12 h in all the assays. In conclusion, AC should be taken into consideration when interpreting in vitro results as their negative effect on bacterial growth may be mistakenly interpreted as HIP efficacy. ACD and Li Heparin appear more appropriate for the selected assays.
Collapse
|
6
|
Bobe JR, Jutras BL, Horn EJ, Embers ME, Bailey A, Moritz RL, Zhang Y, Soloski MJ, Ostfeld RS, Marconi RT, Aucott J, Ma'ayan A, Keesing F, Lewis K, Ben Mamoun C, Rebman AW, McClune ME, Breitschwerdt EB, Reddy PJ, Maggi R, Yang F, Nemser B, Ozcan A, Garner O, Di Carlo D, Ballard Z, Joung HA, Garcia-Romeu A, Griffiths RR, Baumgarth N, Fallon BA. Recent Progress in Lyme Disease and Remaining Challenges. Front Med (Lausanne) 2021; 8:666554. [PMID: 34485323 PMCID: PMC8416313 DOI: 10.3389/fmed.2021.666554] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/12/2021] [Indexed: 12/14/2022] Open
Abstract
Lyme disease (also known as Lyme borreliosis) is the most common vector-borne disease in the United States with an estimated 476,000 cases per year. While historically, the long-term impact of Lyme disease on patients has been controversial, mounting evidence supports the idea that a substantial number of patients experience persistent symptoms following treatment. The research community has largely lacked the necessary funding to properly advance the scientific and clinical understanding of the disease, or to develop and evaluate innovative approaches for prevention, diagnosis, and treatment. Given the many outstanding questions raised into the diagnosis, clinical presentation and treatment of Lyme disease, and the underlying molecular mechanisms that trigger persistent disease, there is an urgent need for more support. This review article summarizes progress over the past 5 years in our understanding of Lyme and tick-borne diseases in the United States and highlights remaining challenges.
Collapse
Affiliation(s)
- Jason R. Bobe
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Brandon L. Jutras
- Department of Biochemistry, Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
| | | | - Monica E. Embers
- Tulane University Health Sciences, New Orleans, LA, United States
| | - Allison Bailey
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Ying Zhang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mark J. Soloski
- Division of Rheumatology, Department of Medicine, Lyme Disease Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | | | - Richard T. Marconi
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, VA, United States
| | - John Aucott
- Division of Rheumatology, Department of Medicine, Lyme Disease Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Avi Ma'ayan
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Kim Lewis
- Department of Biology, Northeastern University, Boston, MA, United States
| | | | - Alison W. Rebman
- Division of Rheumatology, Department of Medicine, Lyme Disease Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mecaila E. McClune
- Department of Biochemistry, Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
| | - Edward B. Breitschwerdt
- Department of Clinical Sciences, Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | | | - Ricardo Maggi
- Department of Clinical Sciences, Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Frank Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Bennett Nemser
- Steven & Alexandra Cohen Foundation, Stamford, CT, United States
| | - Aydogan Ozcan
- University of California, Los Angeles, Los Angeles, CA, United States
| | - Omai Garner
- University of California, Los Angeles, Los Angeles, CA, United States
| | - Dino Di Carlo
- University of California, Los Angeles, Los Angeles, CA, United States
| | - Zachary Ballard
- University of California, Los Angeles, Los Angeles, CA, United States
| | - Hyou-Arm Joung
- University of California, Los Angeles, Los Angeles, CA, United States
| | - Albert Garcia-Romeu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Roland R. Griffiths
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nicole Baumgarth
- Center for Immunology and Infectious Diseases and the Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Brian A. Fallon
- Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
7
|
Erdei A, Kovács KG, Nagy-Baló Z, Lukácsi S, Mácsik-Valent B, Kurucz I, Bajtay Z. New aspects in the regulation of human B cell functions by complement receptors CR1, CR2, CR3 and CR4. Immunol Lett 2021; 237:42-57. [PMID: 34186155 DOI: 10.1016/j.imlet.2021.06.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 10/21/2022]
Abstract
The involvement of complement in the regulation of antibody responses has been known for long. By now several additional B cell functions - including cytokine production and antigen presentation - have also been shown to be regulated by complement proteins. Most of these important activities are mediated by receptors interacting with activation fragments of the central component of the complement system C3, such as C3b, iC3b and C3d, which are covalently attached to antigens and immune complexes. This review summarizes the role of complement receptors interacting with these ligands, namely CR1 (CD35), CR2 (CD21), CR3 (CD11b/CD18) and CR4 (CD11c/CD18) expressed by B cells in health and disease. Although we focus on human B lymphocytes, we also aim to call the attention to important differences between human and mouse systems.
Collapse
Affiliation(s)
- Anna Erdei
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary; MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary.
| | - Kristóf G Kovács
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Zsuzsa Nagy-Baló
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary
| | - Szilvia Lukácsi
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary
| | | | - István Kurucz
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary
| | - Zsuzsa Bajtay
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary; MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
8
|
Woitzik P, Linder S. Molecular Mechanisms of Borrelia burgdorferi Phagocytosis and Intracellular Processing by Human Macrophages. BIOLOGY 2021; 10:567. [PMID: 34206480 PMCID: PMC8301104 DOI: 10.3390/biology10070567] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/16/2021] [Accepted: 06/20/2021] [Indexed: 12/21/2022]
Abstract
Lyme disease is the most common vector-borne illness in North America and Europe. Its causative agents are spirochetes of the Borrelia burgdorferi sensu latu complex. Infection with borreliae can manifest in different tissues, most commonly in the skin and joints, but in severe cases also in the nervous systems and the heart. The immune response of the host is a crucial factor for preventing the development or progression of Lyme disease. Macrophages are part of the innate immune system and thus one of the first cells to encounter infecting borreliae. As professional phagocytes, they are capable of recognition, uptake, intracellular processing and final elimination of borreliae. This sequence of events involves the initial capture and internalization by actin-rich cellular protrusions, filopodia and coiling pseudopods. Uptake into phagosomes is followed by compaction of the elongated spirochetes and degradation in mature phagolysosomes. In this review, we discuss the current knowledge about the processes and molecular mechanisms involved in recognition, capturing, uptake and intracellular processing of Borrelia by human macrophages. Moreover, we highlight interactions between macrophages and other cells of the immune system during these processes and point out open questions in the intracellular processing of borreliae, which include potential escape strategies of Borrelia.
Collapse
Affiliation(s)
| | - Stefan Linder
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, 20246 Hamburg, Germany;
| |
Collapse
|
9
|
Benjamin SJ, Hawley KL, Vera-Licona P, La Vake CJ, Cervantes JL, Ruan Y, Radolf JD, Salazar JC. Macrophage mediated recognition and clearance of Borrelia burgdorferi elicits MyD88-dependent and -independent phagosomal signals that contribute to phagocytosis and inflammation. BMC Immunol 2021; 22:32. [PMID: 34000990 PMCID: PMC8127205 DOI: 10.1186/s12865-021-00418-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 04/22/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Macrophages play prominent roles in bacteria recognition and clearance, including Borrelia burgdorferi (Bb), the Lyme disease spirochete. To elucidate mechanisms by which MyD88/TLR signaling enhances clearance of Bb by macrophages, we studied wildtype (WT) and MyD88-/- Bb-stimulated bone marrow-derived macrophages (BMDMs). RESULTS MyD88-/- BMDMs exhibit impaired uptake of spirochetes but comparable maturation of phagosomes following internalization of spirochetes. RNA-sequencing of infected WT and MyD88-/- BMDMs identified a large cohort of differentially expressed MyD88-dependent genes associated with re-organization of actin and cytoskeleton during phagocytosis along with several MyD88-independent chemokines involved in inflammatory cell recruitment. We computationally generated networks which identified several MyD88-dependent intermediate proteins (Rhoq and Cyfip1) that are known to mediate inflammation and phagocytosis respectively. CONCLUSION Our findings show that MyD88 signaling enhances, but is not required, for bacterial uptake or phagosomal maturation and provide mechanistic insights into how MyD88-mediated phagosomal signaling enhances Bb uptake and clearance.
Collapse
Affiliation(s)
- Sarah J Benjamin
- Department of Pediatrics, UConn Health, Farmington, CT, 06030, USA
- Department of Immunology, UConn Health, Farmington, CT, 06030, USA
| | - Kelly L Hawley
- Department of Pediatrics, UConn Health, Farmington, CT, 06030, USA
- Division of Infectious Diseases, Connecticut Children's, Hartford, CT, 06106, USA
| | - Paola Vera-Licona
- Department of Pediatrics, UConn Health, Farmington, CT, 06030, USA
- Center for Quantitative Medicine, UConn Health, Farmington, CT, 06030, USA
- Department of Cell Biology, UConn Health, Farmington, CT, 06030, USA
- Institute of Systems Genomics, UConn Health, Farmington, CT, 06030, USA
| | - Carson J La Vake
- Department of Pediatrics, UConn Health, Farmington, CT, 06030, USA
| | - Jorge L Cervantes
- Department of Pediatrics, UConn Health, Farmington, CT, 06030, USA
- Division of Infectious Diseases, Connecticut Children's, Hartford, CT, 06106, USA
- Present Address: Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, 79905, USA
| | - Yijun Ruan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Justin D Radolf
- Department of Pediatrics, UConn Health, Farmington, CT, 06030, USA
- Department of Immunology, UConn Health, Farmington, CT, 06030, USA
- Department of Medicine, UConn Health, Farmington, CT, 06030, USA
- Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT, 06030, USA
- Department of Genetics and Genomic Sciences, UConn Health, Farmington, CT, 06030, USA
| | - Juan C Salazar
- Department of Pediatrics, UConn Health, Farmington, CT, 06030, USA.
- Department of Immunology, UConn Health, Farmington, CT, 06030, USA.
- Division of Infectious Diseases, Connecticut Children's, Hartford, CT, 06106, USA.
- Department of Medicine, UConn Health, Farmington, CT, 06030, USA.
- Division of Pediatric Infectious Diseases and Immunology, Connecticut Children's, 282 Washington Street, Hartford, CT, 06106, USA.
| |
Collapse
|
10
|
Owen AM, Fults JB, Patil NK, Hernandez A, Bohannon JK. TLR Agonists as Mediators of Trained Immunity: Mechanistic Insight and Immunotherapeutic Potential to Combat Infection. Front Immunol 2021; 11:622614. [PMID: 33679711 PMCID: PMC7930332 DOI: 10.3389/fimmu.2020.622614] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/24/2020] [Indexed: 12/18/2022] Open
Abstract
Despite advances in critical care medicine, infection remains a significant problem that continues to be complicated with the challenge of antibiotic resistance. Immunocompromised patients are highly susceptible to development of severe infection which often progresses to the life-threatening condition of sepsis. Thus, immunotherapies aimed at boosting host immune defenses are highly attractive strategies to ward off infection and protect patients. Recently there has been mounting evidence that activation of the innate immune system can confer long-term functional reprogramming whereby innate leukocytes mount more robust responses upon secondary exposure to a pathogen for more efficient clearance and host protection, termed trained immunity. Toll-like receptor (TLR) agonists are a class of agents which have been shown to trigger the phenomenon of trained immunity through metabolic reprogramming and epigenetic modifications which drive profound augmentation of antimicrobial functions. Immunomodulatory TLR agonists are also highly beneficial as vaccine adjuvants. This review provides an overview on TLR signaling and our current understanding of TLR agonists which show promise as immunotherapeutic agents for combating infection. A brief discussion on our current understanding of underlying mechanisms is also provided. Although an evolving field, TLR agonists hold strong therapeutic potential as immunomodulators and merit further investigation for clinical translation.
Collapse
Affiliation(s)
- Allison M Owen
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jessica B Fults
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States.,University of Texas Southwestern Medical School, Dallas, TX, United States
| | - Naeem K Patil
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Antonio Hernandez
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Julia K Bohannon
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
11
|
Klose M, Scheungrab M, Luckner M, Wanner G, Linder S. FIB-SEM-based analysis of Borrelia intracellular processing by human macrophages. J Cell Sci 2021; 134:jcs252320. [PMID: 33380490 DOI: 10.1242/jcs.252320] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 12/15/2020] [Indexed: 01/04/2023] Open
Abstract
Borrelia burgdorferi is the causative agent of Lyme disease, a multisystemic disorder affecting primarily skin, joints and nervous system. Successful internalization and intracellular processing of borreliae by immune cells, like macrophages, is decisive for the outcome of a respective infection. Here, we use, for the first time, focused ion beam scanning electron microscopy tomography (FIB-SEM tomography) to visualize the interaction of borreliae with primary human macrophages with high resolution. We report that interaction between macrophages and the elongated and highly motile borreliae can lead to formation of membrane tunnels that extend deeper into the host cytoplasm than the actual phagosome, most probably as a result of partial extrication of captured borreliae. We also show that membrane tubulation at borreliae-containing phagosomes, a process suggested earlier as a mechanism leading to phagosome compaction but hard to visualize in live-cell imaging, is apparently a frequent phenomenon. Finally, we demonstrate that the endoplasmic reticulum (ER) forms multiple STIM1-positive contact sites with both membrane tunnels and phagosome tubulations, confirming the important role of the ER during uptake and intracellular processing of borreliae.
Collapse
Affiliation(s)
- Matthias Klose
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, 20246 Hamburg, Germany
| | | | - Manja Luckner
- Biozentrum der Ludwig-Maximilians-Universität, 82152 Planegg-Martinsried, Germany
| | - Gerhard Wanner
- Biozentrum der Ludwig-Maximilians-Universität, 82152 Planegg-Martinsried, Germany
| | - Stefan Linder
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
12
|
Barriales D, Martín-Ruiz I, Carreras-González A, Montesinos-Robledo M, Azkargorta M, Iloro I, Escobés I, Martín-Mateos T, Atondo E, Palacios A, Gonzalez-Lopez M, Bárcena L, Cortázar AR, Cabrera D, Peña-Cearra A, van Liempd SM, Falcón-Pérez JM, Pascual-Itoiz MA, Flores JM, Abecia L, Pellon A, Martínez-Chantar ML, Aransay AM, Pascual A, Elortza F, Berra E, Lavín JL, Rodríguez H, Anguita J. Borrelia burgdorferi infection induces long-term memory-like responses in macrophages with tissue-wide consequences in the heart. PLoS Biol 2021; 19:e3001062. [PMID: 33395408 PMCID: PMC7808612 DOI: 10.1371/journal.pbio.3001062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 01/14/2021] [Accepted: 12/22/2020] [Indexed: 11/19/2022] Open
Abstract
Lyme carditis is an extracutaneous manifestation of Lyme disease characterized by episodes of atrioventricular block of varying degrees and additional, less reported cardiomyopathies. The molecular changes associated with the response to Borrelia burgdorferi over the course of infection are poorly understood. Here, we identify broad transcriptomic and proteomic changes in the heart during infection that reveal a profound down-regulation of mitochondrial components. We also describe the long-term functional modulation of macrophages exposed to live bacteria, characterized by an augmented glycolytic output, increased spirochetal binding and internalization, and reduced inflammatory responses. In vitro, glycolysis inhibition reduces the production of tumor necrosis factor (TNF) by memory macrophages, whereas in vivo, it produces the reversion of the memory phenotype, the recovery of tissue mitochondrial components, and decreased inflammation and spirochetal burdens. These results show that B. burgdorferi induces long-term, memory-like responses in macrophages with tissue-wide consequences that are amenable to be manipulated in vivo. Lyme carditis is a manifestation of Lyme disease characterized by episodes of atrioventricular block and additional cardiomyopathies. This study describes the proteomic and transcriptomic changes in the heart upon infection with Borrelia burgdorferi, and identifies innate immune memory hallmarks specific to the response to the spirochete that are amenable to therapeutic manipulation.
Collapse
Affiliation(s)
- Diego Barriales
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Itziar Martín-Ruiz
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Ana Carreras-González
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Marta Montesinos-Robledo
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Mikel Azkargorta
- Proteomics Platform, ProteoRed-ISCIII, CIC bioGUNE-BRTA, Derio, Spain
| | - Ibon Iloro
- Proteomics Platform, ProteoRed-ISCIII, CIC bioGUNE-BRTA, Derio, Spain
| | - Iraide Escobés
- Proteomics Platform, ProteoRed-ISCIII, CIC bioGUNE-BRTA, Derio, Spain
| | - Teresa Martín-Mateos
- Physiopathology of the Hypoxia-Signaling Pathway Laboratory, CIC bioGUNE-BRTA, Derio, Spain
| | - Estibaliz Atondo
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Ainhoa Palacios
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | | | - Laura Bárcena
- Genomic Analysis Platform, CIC bioGUNE-BRTA, Derio, Spain
| | | | - Diana Cabrera
- Metabolomics Platform, CIC bioGUNE-BRTA, Derio, Spain
| | - Ainize Peña-Cearra
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | | | - Juan M. Falcón-Pérez
- Metabolomics Platform, CIC bioGUNE-BRTA, Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Miguel A. Pascual-Itoiz
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Juana María Flores
- Department of Animal Medicine and Surgery, Veterinary Faculty, Universidad Complutense de Madrid, Madrid, Spain
| | - Leticia Abecia
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Aize Pellon
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | | | - Ana M. Aransay
- Genomic Analysis Platform, CIC bioGUNE-BRTA, Derio, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Pascual
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Felix Elortza
- Proteomics Platform, ProteoRed-ISCIII, CIC bioGUNE-BRTA, Derio, Spain
| | - Edurne Berra
- Physiopathology of the Hypoxia-Signaling Pathway Laboratory, CIC bioGUNE-BRTA, Derio, Spain
| | | | - Héctor Rodríguez
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
| | - Juan Anguita
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- * E-mail:
| |
Collapse
|
13
|
Bockenstedt LK, Wooten RM, Baumgarth N. Immune Response to Borrelia: Lessons from Lyme Disease Spirochetes. Curr Issues Mol Biol 2020; 42:145-190. [PMID: 33289684 PMCID: PMC10842262 DOI: 10.21775/cimb.042.145] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The mammalian host responds to infection with Borrelia spirochetes through a highly orchestrated immune defense involving innate and adaptive effector functions aimed toward limiting pathogen burdens, minimizing tissue injury, and preventing subsequent reinfection. The evolutionary adaptation of Borrelia spirochetes to their reservoir mammalian hosts may allow for its persistence despite this immune defense. This review summarizes our current understanding of the host immune response to B. burgdorferi sensu lato, the most widely studied Borrelia spp. and etiologic agent of Lyme borreliosis. Pertinent literature will be reviewed with emphasis on in vitro, ex vivo and animal studies that influenced our understanding of both the earliest responses to B. burgdorferi as it enters the mammalian host and those that evolve as spirochetes disseminate and establish infection in multiple tissues. Our focus is on the immune response of inbred mice, the most commonly studied animal model of B. burgdorferi infection and surrogate for one of this pathogen's principle natural reservoir hosts, the white-footed deer mouse. Comparison will be made to the immune responses of humans with Lyme borreliosis. Our goal is to provide an understanding of the dynamics of the mammalian immune response during infection with B. burgdorferi and its relation to the outcomes in reservoir (mouse) and non-reservoir (human) hosts.
Collapse
Affiliation(s)
- Linda K. Bockenstedt
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520-8031, USA
| | - R. Mark Wooten
- Department of Medical Microbiology and Immunology, University of Toledo Health Science Campus, Toledo, OH 43614, USA
| | - Nicole Baumgarth
- Center for Immunology and Infectious Diseases and Dept. Pathology, Microbiology and Immunology, University of California, Davis, Davis CA 95616, USA
| |
Collapse
|
14
|
A combined transcriptomic approach to identify candidates for an anti-tick vaccine blocking B. afzelii transmission. Sci Rep 2020; 10:20061. [PMID: 33208766 PMCID: PMC7674437 DOI: 10.1038/s41598-020-76268-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 09/17/2020] [Indexed: 12/11/2022] Open
Abstract
Ixodes ricinus is the vector for Borrelia afzelii, the predominant cause of Lyme borreliosis in Europe, whereas Ixodes scapularis is the vector for Borrelia burgdorferi in the USA. Transcription of several I. scapularis genes changes in the presence of B. burgdorferi and contributes to successful infection. To what extend B. afzelii influences gene expression in I. ricinus salivary glands is largely unknown. Therefore, we measured expression of uninfected vs. infected tick salivary gland genes during tick feeding using Massive Analysis of cDNA Ends (MACE) and RNAseq, quantifying 26.179 unique transcripts. While tick feeding was the main differentiator, B. afzelii infection significantly affected expression of hundreds of transcripts, including 465 transcripts after 24 h of tick feeding. Validation of the top-20 B. afzelii-upregulated transcripts at 24 h of tick feeding in ten biological genetic distinct replicates showed that expression varied extensively. Three transcripts could be validated, a basic tail protein, a lipocalin and an ixodegrin, and might be involved in B. afzelii transmission. However, vaccination with recombinant forms of these proteins only marginally altered B. afzelii infection in I. ricinus-challenged mice for one of the proteins. Collectively, our data show that identification of tick salivary genes upregulated in the presence of pathogens could serve to identify potential pathogen-blocking vaccine candidates.
Collapse
|
15
|
Constitutive immune mechanisms: mediators of host defence and immune regulation. Nat Rev Immunol 2020; 21:137-150. [PMID: 32782357 PMCID: PMC7418297 DOI: 10.1038/s41577-020-0391-5] [Citation(s) in RCA: 169] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2020] [Indexed: 02/07/2023]
Abstract
The immune system enables organisms to combat infections and to eliminate endogenous challenges. Immune responses can be evoked through diverse inducible pathways. However, various constitutive mechanisms are also required for immunocompetence. The inducible responses of pattern recognition receptors of the innate immune system and antigen-specific receptors of the adaptive immune system are highly effective, but they also have the potential to cause extensive immunopathology and tissue damage, as seen in many infectious and autoinflammatory diseases. By contrast, constitutive innate immune mechanisms, including restriction factors, basal autophagy and proteasomal degradation, tend to limit immune responses, with loss-of-function mutations in these pathways leading to inflammation. Although they function through a broad and heterogeneous set of mechanisms, the constitutive immune responses all function as early barriers to infection and aim to minimize any disruption of homeostasis. Supported by recent human and mouse data, in this Review we compare and contrast the inducible and constitutive mechanisms of immunosurveillance.
Collapse
|
16
|
Distinct Contributions of CD18 Integrins for Binding and Phagocytic Internalization of Pseudomonas aeruginosa. Infect Immun 2020; 88:IAI.00011-20. [PMID: 32041787 DOI: 10.1128/iai.00011-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/06/2020] [Indexed: 01/20/2023] Open
Abstract
Phagocytosis is the key mechanism for host control of Pseudomonas aeruginosa, a motile Gram-negative, opportunistic bacterial pathogen which frequently undergoes adaptation and selection for traits that are advantageous for survival. One such clinically relevant adaptation is the loss of bacterial motility, observed within chronic infections, that is associated with increased antibiotic tolerance and phagocytic resistance. Previous studies using phagocytes from a leukocyte adhesion deficiency type 1 (LAD-I) patient identified CD18 as a putative cell surface receptor for uptake of live P. aeruginosa However, how bacterial motility alters direct engagement with CD18-containing integrins remains unknown. Here we demonstrate, with the use of motile and isogenic nonmotile deletion mutants of two independent strains of P. aeruginosa and with CRISPR-generated CD18-deficient cell lines in human monocytes and murine neutrophils, that CD18 expression facilitates the uptake of both motile and nonmotile P. aeruginosa However, unexpectedly, mechanistic studies revealed that CD18 expression was dispensable for the initial attachment of the bacteria to the host cells, which was validated with ectopic expression of complement receptor 3 (CR3) by CHO cells. Our data support that surface N-linked glycan chains (N-glycans) likely facilitate the initial interaction of bacteria with monocytes and cooperate with CD18 integrins in trans to promote internalization of bacteria. Moreover, talin-1 and kindlin-3 proteins promote uptake, but not binding, of P. aeruginosa by murine neutrophils, which supports a role for CD18 integrin signaling in this process. These findings provide novel insights into the cellular determinants for phagocytic recognition and uptake of P. aeruginosa.
Collapse
|
17
|
Lukácsi S, Mácsik-Valent B, Nagy-Baló Z, Kovács KG, Kliment K, Bajtay Z, Erdei A. Utilization of complement receptors in immune cell-microbe interaction. FEBS Lett 2020; 594:2695-2713. [PMID: 31989596 DOI: 10.1002/1873-3468.13743] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/19/2022]
Abstract
The complement system is a major humoral component of immunity and is essential for the fast elimination of pathogens invading the body. In addition to its indispensable role in innate immunity, the complement system is also involved in pathogen clearance during the effector phase of adaptive immunity. The fastest way of killing the invader is lysis by the membrane attack complex, which is formed by the terminal components of the complement cascade. Not all pathogens are lysed however and, if opsonized by a variety of molecules, they undergo phagocytosis and disposal inside immune cells. The most important complement-derived opsonins are C1q, the first component of the classical pathway, MBL, the initiator of the lectin pathway and C3-derived activation fragments, including C3b, iC3b and C3d, which all serve as ligands for their corresponding receptors. In this review, we discuss how complement receptors are utilized by various immune cells to tackle invading microbes, or by pathogens to evade host response.
Collapse
Affiliation(s)
- Szilvia Lukácsi
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary
| | | | - Zsuzsa Nagy-Baló
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Kristóf G Kovács
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | | | - Zsuzsa Bajtay
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary.,Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Anna Erdei
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary.,Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
18
|
Carreras-González A, Barriales D, Palacios A, Montesinos-Robledo M, Navasa N, Azkargorta M, Peña-Cearra A, Tomás-Cortázar J, Escobes I, Pascual-Itoiz MA, Hradiská J, Kopecký J, Gil-Carton D, Prados-Rosales R, Abecia L, Atondo E, Martín I, Pellón A, Elortza F, Rodríguez H, Anguita J. Regulation of macrophage activity by surface receptors contained within Borrelia burgdorferi-enriched phagosomal fractions. PLoS Pathog 2019; 15:e1008163. [PMID: 31738806 PMCID: PMC6886865 DOI: 10.1371/journal.ppat.1008163] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/02/2019] [Accepted: 10/29/2019] [Indexed: 11/19/2022] Open
Abstract
Macrophages mediate the elimination of pathogens by phagocytosis resulting in the activation of specific signaling pathways that lead to the production of cytokines, chemokines and other factors. Borrelia burgdorferi, the causative agent of Lyme disease, causes a wide variety of pro-inflammatory symptoms. The proinflammatory capacity of macrophages is intimately related to the internalization of the spirochete. However, most receptors mediating this process are largely unknown. We have applied a multiomic approach, including the proteomic analysis of B. burgdorferi-containing phagosome-enriched fractions, to identify surface receptors that are involved in the phagocytic capacity of macrophages as well as their inflammatory output. Sucrose gradient protein fractions of human monocyte-derived macrophages exposed to B. burgdorferi contained the phagocytic receptor, CR3/CD14 highlighting the major role played by these proteins in spirochetal phagocytosis. Other proteins identified in these fractions include C-type lectins, scavenger receptors or Siglecs, of which some are directly involved in the interaction with the spirochete. We also identified the Fc gamma receptor pathway, including the binding receptor, CD64, as involved both in the phagocytosis of, and TNF induction in response to B. burgdorferi in the absence of antibodies. The common gamma chain, FcγR, mediates the phagocytosis of the spirochete, likely through Fc receptors and C-type lectins, in a process that involves Syk activation. Overall, these findings highlight the complex array of receptors involved in the phagocytic response of macrophages to B. burgdorferi. Macrophages eliminate infecting microorganisms through the concerted action of surface receptors and signaling molecules. As a consequence, these cells produce a series of soluble factors that participate in the inflammatory response during infections. The composition of the full complement of receptors that participate in the recognition and internalization of the causative agent of Lyme disease, Borrelia burgdorferi, is largely unknown. We have analyzed the protein composition of phagosomes containing B. burgdorferi from human macrophages and identified a series of surface proteins that may be involved in the process. Through the use of gene silencing techniques, we have determined the participation of several of these receptors both in the internalization of the bacterium and the subsequent inflammatory response. Among these, we have identified the Fc gamma receptor pathway as involved in this process in the absence of antibodies. We have also identified receptors that are directly involved in the attachment of B. burgdorferi, while others seem to have an accessory role in the internalization and/or induction of proinflammatory cytokines in response to the spirochete. These data clarify the complex array of interactions between macrophages and B. burgdorferi and shed light on the overall response to this infectious agent.
Collapse
Affiliation(s)
- Ana Carreras-González
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Diego Barriales
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Ainhoa Palacios
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | | | - Nicolás Navasa
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Mikel Azkargorta
- Proteomics Platform, CIBERehd, ProteoRed-ISCIII, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Ainize Peña-Cearra
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Julen Tomás-Cortázar
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Iraide Escobes
- Proteomics Platform, CIBERehd, ProteoRed-ISCIII, CIC bioGUNE, Derio, Bizkaia, Spain
| | | | - Jana Hradiská
- Faculty of Science, University of South Bohemia, Branišovská, České Budějovice, Czech Republic
| | - Jan Kopecký
- Faculty of Science, University of South Bohemia, Branišovská, České Budějovice, Czech Republic
| | | | - Rafael Prados-Rosales
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Leticia Abecia
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Estíbaliz Atondo
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Itziar Martín
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Aize Pellón
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Félix Elortza
- Proteomics Platform, CIBERehd, ProteoRed-ISCIII, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Héctor Rodríguez
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
| | - Juan Anguita
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE, Derio, Bizkaia, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Bizkaia, Spain
- * E-mail:
| |
Collapse
|
19
|
Klose M, Salloum JE, Gonschior H, Linder S. SNX3 drives maturation of Borrelia phagosomes by forming a hub for PI(3)P, Rab5a, and galectin-9. J Cell Biol 2019; 218:3039-3059. [PMID: 31337623 PMCID: PMC6719455 DOI: 10.1083/jcb.201812106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 05/06/2019] [Accepted: 06/19/2019] [Indexed: 12/17/2022] Open
Abstract
Borrelia burgdorferi is the causative agent of Lyme disease. Klose et al. show that SNX3 drives processing of internalized B. burgdorferi by binding PI(3)P on the phagosome surface and recruiting galectin-9 vesicles, thus forming a convergence point for the endosomal recycling machinery during processing of spirochetes. The spirochete Borrelia burgdorferi, the causative agent of Lyme disease, is internalized by macrophages and processed in phagolysosomes. Phagosomal compaction, a crucial step in phagolysosome maturation, is driven by contact of Rab5a-positive vesicles with the phagosomal coat. We show that the sorting nexin SNX3 is transported with Rab5a vesicles and that its PX domain enables vesicle–phagosome contact by binding to PI(3)P in the phagosomal coat. Moreover, the C-terminal region of SNX3 recruits galectin-9, a lectin implicated in protein and membrane recycling, which we identify as a further regulator of phagosome compaction. SNX3 thus forms a hub for two distinct vesicle populations, constituting a convergence point for the endosomal recycling machinery, to contribute to phagosome maturation and intracellular processing of borreliae. These data also suggest that the helical shape of B. burgdorferi itself, providing sites of high curvature and thus local PI(3)P enrichment at phagosomes, may be one of the driving elements underlying the efficient elimination of spirochetes by immune cells.
Collapse
Affiliation(s)
- Matthias Klose
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| | - Johann E Salloum
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| | | | - Stefan Linder
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| |
Collapse
|
20
|
Lei Q, Li L, Huang W, Qin B, Zhang S. HEV ORF3 downregulatesCD14 and CD64 to impair macrophages phagocytosis through inhibiting JAK/STAT pathway. J Med Virol 2019; 91:1112-1119. [PMID: 30636344 DOI: 10.1002/jmv.25400] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/08/2019] [Indexed: 01/11/2023]
Abstract
Hepatitis E virus (HEV) could induce chronic hepatitis and liver failure with high mortality through unknown mechanisms. The previous study showed that the HEV open reading frames 3 (ORF3) could inhibit macrophages inflammatory response. Impaired macrophages phagocytosis was also found in patients infected with HEV and its nucleic acids could be detected in macrophages. To elucidate the role of HEV ORF3 on phagocytosis, the phagocytosis activation was measured by phagocytosis test, flow cytometry, and phalloidin staining. Meanwhile, the expression of key phagocytic receptors and the activation of transduction pathway were investigated by using reverse transcription real-time quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis. Results of phagocytosis test showed that the HEV ORF3 could significantly impair the absorption capacity of latex beads. Furthermore, results of RT-qPCR and Western blot analysis showed that the expression of CD14 and CD64 decreased. Afterward, the present study showed that the activation of Janus kinase-signal transducer and activator of transcription (JAK/STAT) signaling pathway was inhibited by HEV ORF3 and downregulation of CD14 and CD64 could be reversed by interferon γ, one activator of the JAK1/STAT1 signaling pathway. In conclusion, HEV ORF3 could significantly impair the phagocytosis of macrophage by downregulating expression of CD14 and CD64, which may function by inhibiting the activation of the JAK1/STAT1 signaling pathway.
Collapse
Affiliation(s)
- Qingsong Lei
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Li
- Department of Hepatic Diseases, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Wenxiang Huang
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bo Qin
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shujun Zhang
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
21
|
Navasa N, Fikrig E, Anguita J. Host Defenses to Spirochetes. Clin Immunol 2019. [DOI: 10.1016/b978-0-7020-6896-6.00028-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
22
|
Greenmyer JR, Gaultney RA, Brissette CA, Watt JA. Primary Human Microglia Are Phagocytically Active and Respond to Borrelia burgdorferi With Upregulation of Chemokines and Cytokines. Front Microbiol 2018; 9:811. [PMID: 29922241 PMCID: PMC5996889 DOI: 10.3389/fmicb.2018.00811] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 04/10/2018] [Indexed: 11/13/2022] Open
Abstract
The Lyme disease causing bacterium Borrelia burgdorferi has an affinity for the central nervous system (CNS) and has been isolated from human cerebral spinal fluid by 18 days following Ixodes scapularis tick bite. Signaling from resident immune cells of the CNS could enhance CNS penetration by B. burgdorferi and activated immune cells through the blood brain barrier resulting in multiple neurological complications, collectively termed neuroborreliosis. The ensuing symptoms of neurological impairment likely arise from a glial-driven, host inflammatory response to B. burgdorferi. To date, however, the mechanism by which the bacterium initiates neuroinflammation leading to neural dysfunction remains unclear. We hypothesized that dead B. burgdorferi and bacterial debris persist in the CNS in spite of antibiotic treatment and contribute to the continuing inflammatory response in the CNS. To test our hypothesis, cultures of primary human microglia were incubated with live, antibiotic-killed and antibiotic-killed sonicated B. burgdorferi to define the response of microglia to different forms of the bacterium. We demonstrate that primary human microglia treated with B. burgdorferi show increased expression of pattern recognition receptors and genes known to be involved with cytoskeletal rearrangement and phagocytosis including MARCO, SCARB1, PLA2, PLD2, CD14, and TLR3. In addition, we observed increased expression and secretion of pro-inflammatory mediators and neurotrophic factors such as IL-6, IL-8, CXCL-1, and CXCL-10. Our data also indicate that B. burgdorferi interacts with the cell surface of primary human microglia and may be internalized following this initial interaction. Furthermore, our results indicate that dead and sonicated forms of B. burgdorferi induce a significantly larger inflammatory response than live bacteria. Our results support our hypothesis and provide evidence that microglia contribute to the damaging inflammatory events associated with neuroborreliosis.
Collapse
Affiliation(s)
- Jacob R. Greenmyer
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| | | | - Catherine A. Brissette
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| | - John A. Watt
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States
| |
Collapse
|
23
|
A multi-omic analysis reveals the regulatory role of CD180 during the response of macrophages to Borrelia burgdorferi. Emerg Microbes Infect 2018; 7:19. [PMID: 29511161 PMCID: PMC5841238 DOI: 10.1038/s41426-017-0018-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/08/2017] [Accepted: 12/14/2017] [Indexed: 12/21/2022]
Abstract
Macrophages are cells of the innate immune system with the ability to phagocytose and induce a global pattern of responses that depend on several signaling pathways. We have determined the biosignature of murine bone marrow-derived macrophages and human blood monocytes using transcriptomic and proteomic approaches. We identified a common pattern of genes that are transcriptionally regulated and overall indicate that the response to B. burgdorferi involves the interaction of spirochetal antigens with several inflammatory pathways corresponding to primary (triggered by pattern-recognition receptors) and secondary (induced by proinflammatory cytokines) responses. We also show that the Toll-like receptor family member CD180 is downregulated by the stimulation of macrophages, but not monocytes, with the spirochete. Silencing Cd180 results in increased phagocytosis while tempering the production of the proinflammatory cytokine TNF. Cd180-silenced cells produce increased levels of Itgam and surface CD11b, suggesting that the regulation of CD180 by the spirochete initiates a cascade that increases CR3-mediated phagocytosis of the bacterium while repressing the consequent inflammatory response.
Collapse
|
24
|
Dold NM, Zeng Q, Zeng X, Jewell CM. A poly(beta-amino ester) activates macrophages independent of NF-κB signaling. Acta Biomater 2018; 68:168-177. [PMID: 29292166 PMCID: PMC6292427 DOI: 10.1016/j.actbio.2017.12.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/03/2017] [Accepted: 12/22/2017] [Indexed: 12/21/2022]
Abstract
Nucleic acid delivery vehicles are poised to play an important role in delivering gene therapy for vaccines and immunotherapies, and in delivering nucleic acid based adjuvants. A number of common polymeric delivery vehicles used in nucleic acid delivery have recently been shown to interact with immune cells and directly stimulate immunogenic responses, particularly in particle form. Poly(beta-amino esters) were designed for nucleic acid delivery and have demonstrated promising performance in a number of vaccine and therapeutic studies. Yet, little work has characterized the mechanisms by which these polymers activate immune cells. Here we demonstrate that a poly(beta-amino ester) activates antigen presenting cells in soluble and particulate forms, and that these effects are independent of TLR signaling pathways. Moreover, we show the polymers induce activation independent of NF-κB signaling, but do activate IRF, an important innate inflammatory pathway. New knowledge linking physicochemical features of poly(beta-amino esters) or other polymeric carriers to inflammatory mechanisms could support more rational design approaches for vaccines and immunotherapies harnessing these materials. SIGNIFICANCE STATEMENT The last several years have brought exciting work exploring biomaterials as delivery vehicles for immunotherapies, vaccines, and gene therapies. However, a gap remains between the striking finding that many biomaterials exhibit intrinsic immunogenic features, and the specific structural properties that drive these responses. The results in the current study indicate PBAEs cause macrophage activation by pathways that are distinct from pathways activated by common vaccine and immunotherapies components, such as toll-like receptor agonists. Thus, the work reveals new mechanistic details that can be exploited in investigating other materials, and to support more rational design of future biomaterial vaccines and immunotherapy carriers.
Collapse
Affiliation(s)
- Neil M Dold
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA
| | - Qin Zeng
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA
| | - Xiangbin Zeng
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, 8228 Paint Branch Drive, College Park, MD 20742, USA; United States Department of Veterans Affairs, 10 North Greene Street, Baltimore, MD 21201, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201, USA; Marlene and Stewart Greenebaum Cancer Center, 22 South Greene Street, Baltimore, MD 21201, USA.
| |
Collapse
|
25
|
Sanmarco LM, Eberhardt N, Ponce NE, Cano RC, Bonacci G, Aoki MP. New Insights into the Immunobiology of Mononuclear Phagocytic Cells and Their Relevance to the Pathogenesis of Cardiovascular Diseases. Front Immunol 2018; 8:1921. [PMID: 29375564 PMCID: PMC5767236 DOI: 10.3389/fimmu.2017.01921] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/14/2017] [Indexed: 12/18/2022] Open
Abstract
Macrophages are the primary immune cells that reside within the myocardium, suggesting that these mononuclear phagocytes are essential in the orchestration of cardiac immunity and homeostasis. Independent of the nature of the injury, the heart triggers leukocyte activation and recruitment. However, inflammation is harmful to this vital terminally differentiated organ with extremely poor regenerative capacity. As such, cardiac tissue has evolved particular strategies to increase the stress tolerance and minimize the impact of inflammation. In this sense, growing evidences show that mononuclear phagocytic cells are particularly dynamic during cardiac inflammation or infection and would actively participate in tissue repair and functional recovery. They respond to soluble mediators such as metabolites or cytokines, which play central roles in the timing of the intrinsic cardiac stress response. During myocardial infarction two distinct phases of monocyte influx have been identified. Upon infarction, the heart modulates its chemokine expression profile that sequentially and actively recruits inflammatory monocytes, first, and healing monocytes, later. In the same way, a sudden switch from inflammatory macrophages (with microbicidal effectors) toward anti-inflammatory macrophages occurs within the myocardium very shortly after infection with Trypanosoma cruzi, the causal agent of Chagas cardiomyopathy. While in sterile injury, healing response is necessary to stop tissue damage; during an intracellular infection, the anti-inflammatory milieu in infected hearts would promote microbial persistence. The balance of mononuclear phagocytic cells seems to be also dynamic in atherosclerosis influencing plaque initiation and fate. This review summarizes the participation of mononuclear phagocyte system in cardiovascular diseases, keeping in mind that the immune system evolved to promote the reestablishment of tissue homeostasis following infection/injury, and that the effects of different mediators could modulate the magnitude and quality of the immune response. The knowledge of the effects triggered by diverse mediators would serve to identify new therapeutic targets in different cardiovascular pathologies.
Collapse
Affiliation(s)
- Liliana Maria Sanmarco
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Córdoba, Argentina
| | - Natalia Eberhardt
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Córdoba, Argentina
| | - Nicolás Eric Ponce
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Laboratorio de Neuropatología Experimental, Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Roxana Carolina Cano
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Universidad Católica de Córdoba, Unidad Asociada Área Ciencias Agrarias, Ingeniería, Ciencias Biológicas y de la Salud, Facultad de Ciencias Químicas, Córdoba, Argentina
| | - Gustavo Bonacci
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Córdoba, Argentina
| | - Maria Pilar Aoki
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Córdoba, Argentina
| |
Collapse
|
26
|
Non-identical twins: Different faces of CR3 and CR4 in myeloid and lymphoid cells of mice and men. Semin Cell Dev Biol 2017; 85:110-121. [PMID: 29174917 DOI: 10.1016/j.semcdb.2017.11.025] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 01/08/2023]
Abstract
Integrins are cell membrane receptors that are involved in essential physiological and serious pathological processes. Their main role is to ensure a closely regulated link between the extracellular matrix and the intracellular cytoskeletal network enabling cells to react to environmental stimuli. Complement receptor type 3 (CR3, αMβ2, CD11b/CD18) and type 4 (CR4, αXβ2, CD11c/CD18) are members of the β2-integrin family expressed on most white blood cells. Both receptors bind multiple ligands like iC3b, ICAM, fibrinogen or LPS. β2-integrins are accepted to play important roles in cellular adhesion, migration, phagocytosis, ECM rearrangement and inflammation. Several pathological conditions are linked to the impaired functions of these receptors. CR3 and CR4 are generally thought to mediate overlapping functions in monocytes, macrophages and dendritic cells, therefore the potential distinctive role of these receptors has not been investigated so far in satisfactory details. Lately it has become clear that a functional segregation has evolved between the two receptors regarding phagocytosis, cellular adhesion and podosome formation. In addition to their tasks on myeloid cells, the expression and function of CR3 and CR4 on lymphocytes have also gained interest recently. The picture is further complicated by the fact that while these β2-integrins are expressed by immune cells both in mice and humans, there are significant differences in their expression level, functions and the pathological consequences of genetic defects. Here we aim to summarize our current knowledge on CR3 and CR4 and highlight the functional differences between these receptors, involving their expression in myeloid and lymphoid cells of both men and mice.
Collapse
|
27
|
Hawley KL, Cruz AR, Benjamin SJ, La Vake CJ, Cervantes JL, LeDoyt M, Ramirez LG, Mandich D, Fiel-Gan M, Caimano MJ, Radolf JD, Salazar JC. IFNγ Enhances CD64-Potentiated Phagocytosis of Treponema pallidum Opsonized with Human Syphilitic Serum by Human Macrophages. Front Immunol 2017; 8:1227. [PMID: 29051759 PMCID: PMC5633599 DOI: 10.3389/fimmu.2017.01227] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 09/15/2017] [Indexed: 12/23/2022] Open
Abstract
Syphilis is a multi-stage, sexually transmitted disease caused by the spirochete Treponema pallidum (Tp). Considered broadly, syphilis can be conceptualized as a dualistic process in which spirochete-driven inflammation, the cause of clinical manifestations, coexists to varying extents with bacterial persistence. Inflammation is elicited in the tissues, along with the persistence of spirochetes to keep driving a robust immune response while evading host defenses; this duality is best exemplified during the florid, disseminated stage called secondary syphilis (SS). SS lesions typically contain copious amounts of spirochetes along with a mixed cellular infiltrate consisting of CD4+ T cells, CD8+ T cells, NK cells, plasma cells, and macrophages. In the rabbit model, Tp are cleared by macrophages via antibody-mediated opsonophagocytosis. Previously, we demonstrated that human syphilitic serum (HSS) promotes efficient uptake of Tp by human monocytes and that opsonophagocytosis of Tp markedly enhances cytokine production. Herein, we used monocyte-derived macrophages to study Tp–macrophage interactions ex vivo. In the absence of HSS, monocyte-derived macrophages internalized low numbers of Tp and secreted little cytokine (e.g., TNF). By contrast, these same macrophages internalized large numbers of unopsonized Borrelia burgdorferi and secreted robust levels of cytokines. Maturation of macrophages with M-CSF and IFNγ resulted in a macrophage phenotype with increased expression of HLA-DR, CD14, inducible nitric oxide synthase, TLR2, TLR8, and the Fcγ receptors (FcγR) CD64 and CD16, even in the absence of LPS. Importantly, IFNγ-polarized macrophages resulted in a statistically significant increase in opsonophagocytosis of Tp accompanied by enhanced production of cytokines, macrophage activation markers (CD40, CD80), TLRs (TLR2, TLR7, TLR8), chemokines (CCL19, CXCL10, CXCL11), and TH1-promoting cytokines (IL-12, IL-15). Finally, the blockade of FcγRs, primarily CD64, significantly diminished spirochetal uptake and proinflammatory cytokine secretion by IFNγ-stimulated macrophages. Our ex vivo studies demonstrate the importance of CD64-potentiated uptake of opsonized Tp and suggest that IFNγ-activated macrophages have an important role in the context of early syphilis. Our study results also provide an ex vivo surrogate system for use in future syphilis vaccine studies.
Collapse
Affiliation(s)
- Kelly L Hawley
- Department of Pediatrics, UConn Health, Farmington, CT, United States.,Division of Infectious Diseases, Connecticut Children's Medical Center, Hartford, CT, United States
| | - Adriana R Cruz
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
| | - Sarah J Benjamin
- Department of Pediatrics, UConn Health, Farmington, CT, United States.,Division of Infectious Diseases, Connecticut Children's Medical Center, Hartford, CT, United States.,Department of Immunology, UConn Health, Farmington, CT, United States
| | - Carson J La Vake
- Department of Pediatrics, UConn Health, Farmington, CT, United States
| | - Jorge L Cervantes
- Department of Pediatrics, UConn Health, Farmington, CT, United States.,Division of Infectious Diseases, Connecticut Children's Medical Center, Hartford, CT, United States
| | - Morgan LeDoyt
- Department of Medicine, UConn Health, Farmington, CT, United States
| | - Lady G Ramirez
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
| | - Daniza Mandich
- Department of Pathology, Hartford Hospital, Hartford, CT, United States
| | - Mary Fiel-Gan
- Department of Pathology, Hartford Hospital, Hartford, CT, United States
| | | | - Justin D Radolf
- Department of Pediatrics, UConn Health, Farmington, CT, United States.,Division of Infectious Diseases, Connecticut Children's Medical Center, Hartford, CT, United States.,Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia.,Department of Immunology, UConn Health, Farmington, CT, United States.,Department of Medicine, UConn Health, Farmington, CT, United States.,Department of Genetics and Developmental Biology, UConn Health, Farmington, CT, United States
| | - Juan C Salazar
- Department of Pediatrics, UConn Health, Farmington, CT, United States.,Division of Infectious Diseases, Connecticut Children's Medical Center, Hartford, CT, United States.,Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia.,Department of Immunology, UConn Health, Farmington, CT, United States
| |
Collapse
|
28
|
Phagocytic Receptors Activate Syk and Src Signaling during Borrelia burgdorferi Phagocytosis. Infect Immun 2017; 85:IAI.00004-17. [PMID: 28717031 DOI: 10.1128/iai.00004-17] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/10/2017] [Indexed: 11/20/2022] Open
Abstract
Phagocytosis of the Lyme disease-causing pathogen Borrelia burgdorferi has been shown to be important for generating an inflammatory response to the pathogen. As a result, understanding the mechanisms of phagocytosis has been an area of great interest in the field of Lyme disease. Several cell surface receptors that participate in B. burgdorferi phagocytosis have been reported, including the scavenger receptor MARCO and integrin α3β1. We sought to define the mechanisms by which these receptors mediate phagocytosis and to identify signaling pathways activated downstream of these receptors upon contact with B. burgdorferi We identified both Syk and Src signaling pathways as ones that participate in B. burgdorferi phagocytosis and the resulting cytokine activation. In our studies, we found that both MARCO and integrin β1 play a role in the activation of the Src kinase pathway. However, only integrin β1 participates in the activation of Syk. Interestingly, the integrin activates Syk without the help of the signaling adaptor Dap12 or FcRγ. Thus, we report that multiple pathways participate in B. burgdorferi internalization and that different cell surface receptors act simultaneously in cooperation and independently to mediate phagocytosis.
Collapse
|
29
|
da Silva TA, Zorzetto-Fernandes ALV, Cecílio NT, Sardinha-Silva A, Fernandes FF, Roque-Barreira MC. CD14 is critical for TLR2-mediated M1 macrophage activation triggered by N-glycan recognition. Sci Rep 2017; 7:7083. [PMID: 28765651 PMCID: PMC5539197 DOI: 10.1038/s41598-017-07397-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 06/28/2017] [Indexed: 12/22/2022] Open
Abstract
Agonist interaction with Toll-like receptors (TLRs) induces T cell-mediated immunity, which is effective against intracellular pathogens. Consequently, TLR agonists are being tried as immunomodulatory agents. The lectin ArtinM targets TLR2 N-glycans on macrophages, induces cytokines production, and promotes T helper-1 immunity, a process that culminates in resistance to several parasitic and fungal infections in vivo. Because co-receptors influence agonist binding to TLRs, we investigated whether CD14 is required for macrophage activation induced by ArtinM. Macrophages from wild-type mice stimulated by ArtinM not only produced cytokines but also had the following activation profile: (i) expression of M1 polarization markers; (ii) nitrite oxide production; (iii) cellular migration; (iv) enhanced phagocytic and fungicide activity; (v) modulation of TLR2 expression; and (vi) activation of NF-κB pathway. This activation profile induced by ArtinM was evaluated in macrophages lacking CD14 that showed none of the ArtinM effects. We demonstrated by immunoprecipitation and sugar inhibition assays the physical interaction of ArtinM, TLR2, and CD14, which depends on recognition of the trimannoside that constitutes the core of N-glycans. Thus, our study showed that CD14 is critical for ArtinM-induced macrophage activation, providing fundamental insight into the design of anti-infective therapies based on carbohydrate recognition.
Collapse
Affiliation(s)
- Thiago Aparecido da Silva
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto-USP, Ribeirão Preto, 14049-900, São Paulo, Brazil
| | - André L V Zorzetto-Fernandes
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto-USP, Ribeirão Preto, 14049-900, São Paulo, Brazil
| | - Nerry T Cecílio
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto-USP, Ribeirão Preto, 14049-900, São Paulo, Brazil
| | - Aline Sardinha-Silva
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto-USP, Ribeirão Preto, 14049-900, São Paulo, Brazil
| | - Fabrício Freitas Fernandes
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto-USP, Ribeirão Preto, 14049-900, São Paulo, Brazil
| | - Maria Cristina Roque-Barreira
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto-USP, Ribeirão Preto, 14049-900, São Paulo, Brazil.
| |
Collapse
|
30
|
Zlotnikov N, Javid A, Ahmed M, Eshghi A, Tang TT, Arya A, Bansal A, Matar F, Parikh M, Ebady R, Koh A, Gupta N, Song P, Zhang Y, Newbigging S, Wormser GP, Schwartz I, Inman R, Glogauer M, Moriarty TJ. Infection with the Lyme disease pathogen suppresses innate immunity in mice with diet-induced obesity. Cell Microbiol 2016; 19. [PMID: 27794208 PMCID: PMC5383418 DOI: 10.1111/cmi.12689] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 09/17/2016] [Accepted: 10/24/2016] [Indexed: 12/15/2022]
Abstract
Obesity is a major global public health concern. Immune responses implicated in obesity also control certain infections. We investigated the effects of high‐fat diet‐induced obesity (DIO) on infection with the Lyme disease bacterium Borrelia burgdorferi in mice. DIO was associated with systemic suppression of neutrophil‐ and macrophage‐based innate immune responses. These included bacterial uptake and cytokine production, and systemic, progressive impairment of bacterial clearance, and increased carditis severity. B. burgdorferi‐infected mice fed normal diet also gained weight at the same rate as uninfected mice fed high‐fat diet, toll‐like receptor 4 deficiency rescued bacterial clearance defects, which greater in female than male mice, and killing of an unrelated bacterium (Escherichia coli) by bone marrow‐derived macrophages from obese, B. burgdorferi‐infected mice was also affected. Importantly, innate immune suppression increased with infection duration and depended on cooperative and synergistic interactions between DIO and B. burgdorferi infection. Thus, obesity and B. burgdorferi infection cooperatively and progressively suppressed innate immunity in mice.
Collapse
Affiliation(s)
- Nataliya Zlotnikov
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Ashkan Javid
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Mijhgan Ahmed
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Azad Eshghi
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Tian Tian Tang
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Anoop Arya
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Anil Bansal
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Fatima Matar
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Maitry Parikh
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Rhodaba Ebady
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Adeline Koh
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Nupur Gupta
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Peng Song
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Yang Zhang
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Susan Newbigging
- Mount Sinai Hospital Research Institute/Toronto Centre for Phenogenomics, Toronto, Ontario, Canada
| | - Gary P Wormser
- Division of Infectious Diseases, New York Medical College, New York, USA
| | - Ira Schwartz
- Department of Microbiology and Immunology, New York Medical College, New York, USA
| | - Robert Inman
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto Hospital-Western Division, Toronto, Ontario, Canada
| | - Michael Glogauer
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Tara J Moriarty
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Machacek C, Supper V, Leksa V, Mitulovic G, Spittler A, Drbal K, Suchanek M, Ohradanova-Repic A, Stockinger H. Folate Receptor β Regulates Integrin CD11b/CD18 Adhesion of a Macrophage Subset to Collagen. THE JOURNAL OF IMMUNOLOGY 2016; 197:2229-38. [PMID: 27534550 DOI: 10.4049/jimmunol.1501878] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 07/14/2016] [Indexed: 11/19/2022]
Abstract
Folate, also known as vitamin B9, is necessary for essential cellular functions such as DNA synthesis, repair, and methylation. It is supplied to the cell via several transporters and receptors, including folate receptor (FR) β, a GPI-anchored protein belonging to the folate receptor family. As FRβ shows a restricted expression to cells of myeloid origin and only a subset of activated macrophages and placental cells have been shown to express functional FRβ, it represents a promising target for future therapeutic strategies. In this study, we performed affinity purification and mass spectrometric analysis of the protein microenvironment of FRβ in the plasma membrane of human FRβ(+) macrophages and FRβ-transduced monocytic THP-1 cells. In this manner, we identified a novel role of FRβ: that is, we report functional interactions of FRβ with receptors mediating cellular adhesion, in particular the CD11b/CD18 β2 integrin heterodimer complement receptor type 3/Mac-1. This interaction results in impeded adhesion of FRβ(+) human primary macrophages and THP-1 cells to collagen in comparison with their FRβ(-) counterparts. We further show that FRβ is only expressed by human macrophages when differentiated with M-CSF. These findings thus identify FRβ as a novel CD11b/CD18 regulator for trafficking and homing of a subset of macrophages on collagen.
Collapse
Affiliation(s)
- Christian Machacek
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Verena Supper
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Vladimir Leksa
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia
| | - Goran Mitulovic
- Department of Clinical Chemistry and Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Andreas Spittler
- Department of Surgery and Core Facility Flow Cytometry, Medical University of Vienna, 1090 Vienna, Austria; and
| | | | | | - Anna Ohradanova-Repic
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Hannes Stockinger
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
32
|
Actin-Dependent Regulation of Borrelia burgdorferi Phagocytosis by Macrophages. Curr Top Microbiol Immunol 2016; 399:133-154. [DOI: 10.1007/82_2016_26] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
33
|
Outer surface protein OspC is an antiphagocytic factor that protects Borrelia burgdorferi from phagocytosis by macrophages. Infect Immun 2015; 83:4848-60. [PMID: 26438793 DOI: 10.1128/iai.01215-15] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 09/27/2015] [Indexed: 12/15/2022] Open
Abstract
Outer surface protein C (OspC) is one of the major lipoproteins expressed on the surface of Borrelia burgdorferi during tick feeding and the early phase of mammalian infection. OspC is required for B. burgdorferi to establish infection in both immunocompetent and SCID mice and has been proposed to facilitate evasion of innate immune defenses. However, the exact biological function of OspC remains elusive. In this study, we showed that the ospC-deficient spirochete could not establish infection in NOD-scid IL2rγ(null) mice that lack B cells, T cells, NK cells, and lytic complement. The ospC mutant also could not establish infection in anti-Ly6G-treated SCID and C3H/HeN mice (depletion of neutrophils). However, depletion of mononuclear phagocytes at the skin site of inoculation in SCID and C3H/HeN mice allowed the ospC mutant to establish infection in vivo. In phagocyte-depleted mice, the ospC mutant was able to colonize the joints and triggered neutrophilia during dissemination. Furthermore, we found that phagocytosis of green fluorescent protein (GFP)-expressing ospC mutant spirochetes by murine peritoneal macrophages and human THP-1 macrophage-like cells, but not in PMN-HL60, was significantly higher than parental wild-type B. burgdorferi strains, suggesting that OspC has an antiphagocytic property. In addition, overproduction of OspC in spirochetes also decreased the uptake of spirochetes by murine peritoneal macrophages. Together, our findings provide evidence that mononuclear phagocytes play a key role in clearance of the ospC mutant and that OspC promotes spirochetes' evasion of macrophages during early Lyme borreliosis.
Collapse
|
34
|
Navasa N, Martin-Ruiz I, Atondo E, Sutherland JD, Angel Pascual-Itoiz M, Carreras-González A, Izadi H, Tomás-Cortázar J, Ayaz F, Martin-Martin N, Torres IM, Barrio R, Carracedo A, Olivera ER, Rincón M, Anguita J. Ikaros mediates the DNA methylation-independent silencing of MCJ/DNAJC15 gene expression in macrophages. Sci Rep 2015; 5:14692. [PMID: 26419808 PMCID: PMC4588509 DOI: 10.1038/srep14692] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 09/04/2015] [Indexed: 01/14/2023] Open
Abstract
MCJ (DNAJC15) is a mitochondrial protein that regulates the mitochondrial metabolic status of macrophages and their response to inflammatory stimuli. CpG island methylation in cancer cells constitutes the only mechanism identified for the regulation of MCJ gene expression. However, whether DNA methylation or transcriptional regulation mechanisms are involved in the physiological control of this gene expression in non-tumor cells remains unknown. We now demonstrate a mechanism of regulation of MCJ expression that is independent of DNA methylation. IFNγ, a protective cytokine against cardiac inflammation during Lyme borreliosis, represses MCJ transcription in macrophages. The transcriptional regulator, Ikaros, binds to the MCJ promoter in a Casein kinase II-dependent manner, and mediates the repression of MCJ expression. These results identify the MCJ gene as a transcriptional target of IFNγ and provide evidence of the dynamic adaptation of normal tissues to changes in the environment as a way to adapt metabolically to new conditions.
Collapse
Affiliation(s)
- Nicolás Navasa
- Department of Veterinary and Animal Sciences. University of Massachusetts Amherst. Amherst, MA 01003.,CIC bioGUNE. 48160 Derio, Bizkaia, Spain
| | | | | | | | | | | | - Hooman Izadi
- Department of Veterinary and Animal Sciences. University of Massachusetts Amherst. Amherst, MA 01003
| | | | - Furkan Ayaz
- Department of Veterinary and Animal Sciences. University of Massachusetts Amherst. Amherst, MA 01003
| | | | - Iviana M Torres
- Department of Veterinary and Animal Sciences. University of Massachusetts Amherst. Amherst, MA 01003
| | | | - Arkaitz Carracedo
- CIC bioGUNE. 48160 Derio, Bizkaia, Spain.,Ikerbasque, Basque Foundation for Science. 48011 Bilbao, Bizkaia, Spain.,Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), P. O. Box 644, E-48080 Bilbao, Spain
| | - Elias R Olivera
- Department of Molecular Biology, Veterinary School, University of León. 24071 León, Spain
| | - Mercedes Rincón
- Department of Medicine. University of Vermont College of Medicine. Burlington, VT 05405
| | - Juan Anguita
- Department of Veterinary and Animal Sciences. University of Massachusetts Amherst. Amherst, MA 01003.,CIC bioGUNE. 48160 Derio, Bizkaia, Spain.,Ikerbasque, Basque Foundation for Science. 48011 Bilbao, Bizkaia, Spain
| |
Collapse
|
35
|
Wenzel UA, Fernandez-Santoscoy M, Tam MA, Tegtmeyer P, Wick MJ. Synergy between CD40 and MyD88 Does Not Influence Host Survival to Salmonella Infection. Front Immunol 2015; 6:460. [PMID: 26441965 PMCID: PMC4568434 DOI: 10.3389/fimmu.2015.00460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/24/2015] [Indexed: 01/24/2023] Open
Abstract
Previous studies using purified toll-like receptor (TLR) ligands plus agonistic anti-CD40 antibodies showed that TLRs and CD40 can act synergistically on dendritic cells (DCs) to optimize T cell activation and Th1 differentiation. However, a synergistic effect of TLRs and CD40 during bacterial infection is not known. Here, we show that mice lacking the TLR adaptor MyD88 alone, or lacking both MyD88 and CD40 [double knockout (DKO) mice], are compromised in survival to Salmonella infection but have intact recruitment of neutrophils and inflammatory monocytes as well as unaltered abundance of DC subsets and DC activation in infected tissues. In contrast to infected wildtype and CD40(-/-) mice, both MyD88(-/-) mice and DKO mice lack detectable serum IFN-γ and have elevated IL-10. A synergistic effect of TLRs and CD40 was revealed in co-culture experiments where OT-II T cell proliferation was compromised when DKO DCs were pulsed with OVA protein and OVA323-339 peptide, but not with heat-killed Salmonella expressing OVA (HKSOVA), relative to MyD88(-/-) DCs. By contrast, MyD88(-/-) or DKO DCs pulsed with any of the antigens had a similar ability to induce IFN-γ that was lower than WT or CD40(-/-) DCs. DKO DCs pulsed with HKSOVA, but not with OVA or OVA323-339, had increased IL-10 relative to MyD88(-/-) DCs. Finally, HKSOVA-pulsed MyD88(-/-) and DKO DCs had similar and low induction of NFκB-dependent and -independent genes upon co-culture with OT-II cells. Overall, our data revealed that synergistic effects of CD40 and MyD88 do not influence host survival to Salmonella infection or serum levels of IFN-γ or IL-10. However, synergistic effects of MyD88 and CD40 may be apparent on some (IL-10 production) but not all (OT-II proliferation and IFN-γ production) DC functions and depend on the complexity of the antigen. Indeed, synergistic effects observed using purified ligands and well-defined antigens may not necessarily apply when complex antigens, such as live bacteria, challenge the immune system.
Collapse
Affiliation(s)
- Ulf Alexander Wenzel
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center (MIVAC), Institute of Biomedicine at Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | - Maria Fernandez-Santoscoy
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center (MIVAC), Institute of Biomedicine at Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| | | | - Pia Tegtmeyer
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center (MIVAC), Institute of Biomedicine at Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden ; TWINCORE Centre for Experimental and Clinical Infection Research, Institute for Experimental Infection Research , Hannover , Germany
| | - Mary Jo Wick
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center (MIVAC), Institute of Biomedicine at Sahlgrenska Academy, University of Gothenburg , Gothenburg , Sweden
| |
Collapse
|
36
|
Hawley KL, Olson CM, Carreras-González A, Navasa N, Anguita J. Serum C3 Enhances Complement Receptor 3-Mediated Phagocytosis of Borrelia burgdorferi. Int J Biol Sci 2015; 11:1269-71. [PMID: 26435692 PMCID: PMC4582150 DOI: 10.7150/ijbs.13395] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/03/2015] [Indexed: 11/05/2022] Open
Affiliation(s)
- Kelly L Hawley
- 1. Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 01003 Amherst, MA, USA
| | - Chris M Olson
- 1. Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 01003 Amherst, MA, USA
| | | | - Nicolás Navasa
- 2. CIC bioGUNE, Bizkaia Technology Park, 48160 Derio, Spain
| | - Juan Anguita
- 1. Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, 01003 Amherst, MA, USA ; 2. CIC bioGUNE, Bizkaia Technology Park, 48160 Derio, Spain ; 3. IKERBASQUE, Basque foundation for science, 48013 Bilbao, Spain
| |
Collapse
|
37
|
Naj X, Linder S. ER-Coordinated Activities of Rab22a and Rab5a Drive Phagosomal Compaction and Intracellular Processing of Borrelia burgdorferi by Macrophages. Cell Rep 2015; 12:1816-30. [DOI: 10.1016/j.celrep.2015.08.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 06/24/2015] [Accepted: 08/08/2015] [Indexed: 01/26/2023] Open
|
38
|
McManus M, Cincotta A. Effects of Borrelia on host immune system: Possible consequences for diagnostics. ADVANCES IN INTEGRATIVE MEDICINE 2015. [DOI: 10.1016/j.aimed.2014.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
39
|
Macrophage Polarization during Murine Lyme Borreliosis. Infect Immun 2015; 83:2627-35. [PMID: 25870230 DOI: 10.1128/iai.00369-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 04/09/2015] [Indexed: 12/14/2022] Open
Abstract
Infection of C3H mice with Borrelia burgdorferi, the causative agent of Lyme disease, reliably produces an infectious arthritis and carditis that peak around 3 weeks postinfection and then spontaneously resolve. Macrophage polarization has been suggested to drive inflammation, the clearance of bacteria, and tissue repair and resolution in a variety of infectious disease models. During Lyme disease it is clear that macrophages are capable of clearing Borrelia spirochetes and exhausted neutrophils; however, the role of macrophage phenotype in disease development or resolution has not been studied. Using classical (NOS2) and alternative (CD206) macrophage subset-specific markers, we determined the phenotype of F4/80(+) macrophages within the joints and heart throughout the infection time course. Within the joint, CD206(+) macrophages dominated throughout the course of infection, and NOS2(+) macrophage numbers became elevated only during the peak of inflammation. We also found dual NOS2(+) CD206(+) macrophages which increased during resolution. In contrast to findings for the ankle joints, numbers of NOS2(+) and CD206(+) macrophages in the heart were similar at the peak of inflammation. 5-Lipoxygenase-deficient (5-LOX(-/-)) mice, which display a failure of Lyme arthritis resolution, recruited fewer F4/80(+) cells to the infected joints and heart, but macrophage subset populations were unchanged. These results highlight differences in the inflammatory infiltrates during Lyme arthritis and carditis and demonstrate the coexistence of multiple macrophage subsets within a single inflammatory site.
Collapse
|
40
|
Petnicki-Ocwieja T, Kern A. Mechanisms of Borrelia burgdorferi internalization and intracellular innate immune signaling. Front Cell Infect Microbiol 2014; 4:175. [PMID: 25566512 PMCID: PMC4266086 DOI: 10.3389/fcimb.2014.00175] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 11/27/2014] [Indexed: 01/12/2023] Open
Abstract
Lyme disease is a long-term infection whose most severe pathology is characterized by inflammatory arthritis of the lower bearing joints, carditis, and neuropathy. The inflammatory cascades are initiated through the early recognition of invading Borrelia burgdorferi spirochetes by cells of the innate immune response, such as neutrophils and macrophage. B. burgdorferi does not have an intracellular niche and thus much research has focused on immune pathways activated by pathogen recognition molecules at the cell surface, such as the Toll-like receptors (TLRs). However, in recent years, studies have shown that internalization of the bacterium by host cells is an important component of the defense machinery in response to B. burgdorferi. Upon internalization, B. burgdorferi is trafficked through an endo/lysosomal pathway resulting in the activation of a number of intracellular pathogen recognition receptors including TLRs and Nod-like receptors (NLRs). Here we will review the innate immune molecules that participate in both cell surface and intracellular immune activation by B. burgdorferi.
Collapse
Affiliation(s)
- Tanja Petnicki-Ocwieja
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center Boston, MA, USA
| | - Aurelie Kern
- Division of Geographic Medicine and Infectious Diseases, Tufts Medical Center Boston, MA, USA
| |
Collapse
|
41
|
CD4+ T cells promote antibody production but not sustained affinity maturation during Borrelia burgdorferi infection. Infect Immun 2014; 83:48-56. [PMID: 25312948 DOI: 10.1128/iai.02471-14] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
CD4 T cells are crucial for enhancing B cell-mediated immunity, supporting the induction of high-affinity, class-switched antibody responses, long-lived plasma cells, and memory B cells. Previous studies showed that the immune response to Borrelia burgdorferi appears to lack robust T-dependent B cell responses, as neither long-lived plasma cells nor memory B cells form for months after infection, and nonswitched IgM antibodies are produced continuously during this chronic disease. These data prompted us to evaluate the induction and functionality of B. burgdorferi infection-induced CD4 T(FH) cells. We report that CD4 T cells were effectively primed and T(FH) cells induced after B. burgdorferi infection. These CD4 T cells contributed to the control of B. burgdorferi burden and supported the induction of B. burgdorferi-specific IgG responses. However, while affinity maturation of antibodies against a prototypic T-dependent B. burgdorferi protein, Arthritis-related protein (Arp), were initiated, these increases were reversed later, coinciding with the previously observed involution of germinal centers. The cessation of affinity maturation was not due to the appearance of inhibitory or exhausted CD4 T cells or a strong induction of regulatory T cells. In vitro T-B cocultures demonstrated that T cells isolated from B. burgdorferi-infected but not B. burgdorferi-immunized mice supported the rapid differentiation of B cells into antibody-secreting plasma cells rather than continued proliferation, mirroring the induction of rapid short-lived instead of long-lived T-dependent antibody responses in vivo. The data further suggest that B. burgdorferi infection drives the humoral response away from protective, high-affinity, and long-lived antibody responses and toward the rapid induction of strongly induced, short-lived antibodies of limited efficacy.
Collapse
|
42
|
Sepsis lethality via exacerbated tissue infiltration and TLR-induced cytokine production by neutrophils is integrin α3β1-dependent. Blood 2014; 124:3515-23. [PMID: 25278585 DOI: 10.1182/blood-2014-01-552943] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Integrin-mediated migration of neutrophils to infected tissue sites is vital for pathogen clearance and therefore host survival. Although β2 integrins have been shown to mediate neutrophil transendothelial migration during systemic and local inflammation, relatively little information is available regarding neutrophil migration in sepsis beyond the endothelial cell layer. In this study, we report that integrin α3β1 (VLA-3; CD49c/CD29) is dramatically upregulated on neutrophils isolated from both human septic patients and in mouse models of sepsis. Compared with the α3β1 (low) granulocytes, α3β1 (high) cells from septic animals displayed hyperinflammatory phenotypes. Administration of a α3β1 blocking peptide and conditional deletion of α3 in granulocytes significantly reduced the number of extravasating neutrophils and improved survival in septic mice. In addition, expression of α3β1 on neutrophils was associated with Toll-like receptor-induced inflammatory responses and cytokine productions. Thus, our results show that α3β1 is a novel marker of tissue homing and hyperresponsive neutrophil subtypes in sepsis, and blocking of α3β1 may represent a new therapeutic approach in sepsis treatment.
Collapse
|
43
|
Invariant natural killer T cells act as an extravascular cytotoxic barrier for joint-invading Lyme Borrelia. Proc Natl Acad Sci U S A 2014; 111:13936-41. [PMID: 25205813 DOI: 10.1073/pnas.1404769111] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
CXCR6-GFP(+) cells, which encompass 70% invariant natural killer T cells (iNKT cells), have been found primarily patrolling inside blood vessels in the liver. Although the iNKT cells fail to interact with live pathogens, they do respond to bacterial glycolipids presented by CD1d on liver macrophage that have caught the microbe. In contrast, in this study using dual laser multichannel spinning-disk intravital microscopy of joints, the CXCR6-GFP, which also made up 60-70% iNKT cells, were not found in the vasculature but rather closely apposed to and surrounding the outside of blood vessels, and to a lesser extent throughout the extravascular space. These iNKT cells also differed in behavior, responding rapidly and directly to joint-homing pathogens like Borrelia burgdorferi, which causes Lyme disease. These iNKT cells interacted with B. burgdorferi at the vessel wall and disrupted dissemination attempts by these microbes into joints. Successful penetrance of B. burgdorferi out of the vasculature and into the joint tissue was met by a lethal attack by extravascular iNKT cells through a granzyme-dependent pathway, an observation also made in vitro for iNKT cells from joint but not liver or spleen. These results suggest a novel, critical extravascular iNKT cell immune surveillance in joints that functions as a cytotoxic barrier and explains a large increase in pathogen burden of B. burgdorferi in the joint of iNKT cell-deficient mice, and perhaps the greater susceptibility of humans to this pathogen because of fewer iNKT cells in human joints.
Collapse
|
44
|
Navasa N, Martín I, Iglesias-Pedraz JM, Beraza N, Atondo E, Izadi H, Ayaz F, Fernández-Álvarez S, Hatle K, Som A, Dienz O, Osborne BA, Martínez-Chantar ML, Rincón M, Anguita J. Regulation of oxidative stress by methylation-controlled J protein controls macrophage responses to inflammatory insults. J Infect Dis 2014; 211:135-45. [PMID: 25028693 DOI: 10.1093/infdis/jiu389] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Mitochondria contribute to macrophage immune function through the generation of reactive oxygen species, a byproduct of the mitochondrial respiratory chain. MCJ (also known as DnaJC15) is a mitochondrial inner membrane protein identified as an endogenous inhibitor of respiratory chain complex I. Here we show that MCJ is essential for the production of tumor necrosis factor by macrophages in response to a variety of Toll-like receptor ligands and bacteria, without affecting their phagocytic activity. Loss of MCJ in macrophages results in increased mitochondrial respiration and elevated basal levels of reactive oxygen species that cause activation of the JNK/c-Jun pathway, lead to the upregulation of the TACE (also known as ADAM17) inhibitor TIMP-3, and lead to the inhibition of tumor necrosis factor shedding from the plasma membrane. Consequently, MCJ-deficient mice are resistant to the development of fulminant liver injury upon lipopolysaccharide administration. Thus, attenuation of the mitochondrial respiratory chain by MCJ in macrophages exquisitely regulates the response of macrophages to infectious insults.
Collapse
Affiliation(s)
- Nicolás Navasa
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst Proteomics Unit
| | | | | | | | | | - Hooman Izadi
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst
| | - Furkan Ayaz
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst
| | | | - Ketki Hatle
- Department of Medicine, University of Vermont College of Medicine, Burlington
| | - Abhigyam Som
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst
| | - Oliver Dienz
- Department of Medicine, University of Vermont College of Medicine, Burlington
| | - Barbara A Osborne
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst
| | - Maria Luz Martínez-Chantar
- Metabolomics Unit, CIC bioGUNE Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Technology Park of Bizkaia, Derio Biochemistry and Molecular Biology Department, University of the Basque Country
| | - Mercedes Rincón
- Department of Medicine, University of Vermont College of Medicine, Burlington
| | - Juan Anguita
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst Proteomics Unit Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
45
|
Lochhead RB, Ma Y, Zachary JF, Baltimore D, Zhao JL, Weis JH, O'Connell RM, Weis JJ. MicroRNA-146a provides feedback regulation of lyme arthritis but not carditis during infection with Borrelia burgdorferi. PLoS Pathog 2014; 10:e1004212. [PMID: 24967703 PMCID: PMC4072785 DOI: 10.1371/journal.ppat.1004212] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 05/13/2014] [Indexed: 12/24/2022] Open
Abstract
MicroRNAs have been shown to be important regulators of inflammatory and immune responses and are implicated in several immune disorders including systemic lupus erythematosus and rheumatoid arthritis, but their role in Lyme borreliosis remains unknown. We performed a microarray screen for expression of miRNAs in joint tissue from three mouse strains infected with Borrelia burgdorferi. This screen identified upregulation of miR-146a, a key negative regulator of NF-κB signaling, in all three strains, suggesting it plays an important role in the in vivo response to B. burgdorferi. Infection of B6 miR-146a-/- mice with B. burgdorferi revealed a critical nonredundant role of miR-146a in modulating Lyme arthritis without compromising host immune response or heart inflammation. The impact of miR-146a was specifically localized to the joint, and did not impact lesion development or inflammation in the heart. Furthermore, B6 miR-146a-/- mice had elevated levels of NF-κB-regulated products in joint tissue and serum late in infection. Flow cytometry analysis of various lineages isolated from infected joint tissue of mice showed that myeloid cell infiltration was significantly greater in B6 miR-146a-/- mice, compared to B6, during B. burgdorferi infection. Using bone marrow-derived macrophages, we found that TRAF6, a known target of miR-146a involved in NF-κB activation, was dysregulated in resting and B. burgdorferi-stimulated B6 miR-146a-/- macrophages, and corresponded to elevated IL-1β, IL-6 and CXCL1 production. This dysregulated protein production was also observed in macrophages treated with IL-10 prior to B. burgdorferi stimulation. Peritoneal macrophages from B6 miR-146a-/- mice also showed enhanced phagocytosis of B. burgdorferi. Together, these data show that miR-146a-mediated regulation of TRAF6 and NF-κB, and downstream targets such as IL-1β, IL-6 and CXCL1, are critical for modulation of Lyme arthritis during chronic infection with B. burgdorferi.
Collapse
Affiliation(s)
- Robert B. Lochhead
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Ying Ma
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - James F. Zachary
- Department of Veterinary Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - David Baltimore
- Department of Biology, California Institute of Technology, Pasadena, California, United States of America
| | - Jimmy L. Zhao
- Department of Biology, California Institute of Technology, Pasadena, California, United States of America
| | - John H. Weis
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Ryan M. O'Connell
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Janis J. Weis
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
46
|
Oosting M, Buffen K, van der Meer JWM, Netea MG, Joosten LAB. Innate immunity networks during infection with Borrelia burgdorferi. Crit Rev Microbiol 2014; 42:233-44. [PMID: 24963691 DOI: 10.3109/1040841x.2014.929563] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The recognition of Borrelia species represents a complex process in which multiple components of the immune system are involved. In this review, we summarize the interplay between the host innate system and Borrelia spp., from the recognition by pattern recognition receptors (PRRs) to the induction of a complex network of proinflammatory mediators. Several PRR families are crucial for recognition of Borrelia spp., including Toll-like receptors (TLRs) and Nucleotide Oligomerization Domain (NOD)-like receptors (NLRs). TLR-2 is crucial for the recognition of outer surface protein (Osp)A from Borrelia spp. and together with TLR8 mediates phagocytosis of the microorganism and production of type I interferons. Intracellular receptors such as TLR7, TLR8 and TLR9 on the one hand and the NLR receptor NOD2 on the other hand, represent the second major recognition system of Borrelia. PRR-dependent signals induce the release of pro-inflammatory cytokines such as interleukin-1 and T-helper-derived cytokines, which are thought to mediate the inflammation during Lyme disease. Understanding the regulation of host defense mechanisms against Borrelia has the potential to lead to the discovery of novel immunotherapeutic targets to improve the therapy against Lyme disease.
Collapse
Affiliation(s)
- Marije Oosting
- a Department of Internal Medicine , and.,b Nijmegen Institute of Infection, Inflammation and Immunity (N4i), Radboud University Medical Centre , Nijmegen , The Netherlands
| | - Kathrin Buffen
- a Department of Internal Medicine , and.,b Nijmegen Institute of Infection, Inflammation and Immunity (N4i), Radboud University Medical Centre , Nijmegen , The Netherlands
| | - Jos W M van der Meer
- a Department of Internal Medicine , and.,b Nijmegen Institute of Infection, Inflammation and Immunity (N4i), Radboud University Medical Centre , Nijmegen , The Netherlands
| | - Mihai G Netea
- a Department of Internal Medicine , and.,b Nijmegen Institute of Infection, Inflammation and Immunity (N4i), Radboud University Medical Centre , Nijmegen , The Netherlands
| | - Leo A B Joosten
- a Department of Internal Medicine , and.,b Nijmegen Institute of Infection, Inflammation and Immunity (N4i), Radboud University Medical Centre , Nijmegen , The Netherlands
| |
Collapse
|
47
|
Krupna-Gaylord MA, Liveris D, Love AC, Wormser GP, Schwartz I, Petzke MM. Induction of type I and type III interferons by Borrelia burgdorferi correlates with pathogenesis and requires linear plasmid 36. PLoS One 2014; 9:e100174. [PMID: 24945497 PMCID: PMC4063763 DOI: 10.1371/journal.pone.0100174] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 05/23/2014] [Indexed: 12/28/2022] Open
Abstract
The capacity for Borrelia burgdorferi to cause disseminated infection in humans or mice is associated with the genotype of the infecting strain. The cytokine profiles elicited by B. burgdorferi clinical isolates of different genotype (ribosomal spacer type) groups were assessed in a human PBMC co-incubation model. RST1 isolates, which are more frequently associated with disseminated Lyme disease in humans and mice, induced significantly higher levels of IFN-α and IFN-λ1/IL29 relative to RST3 isolates, which are less frequently associated with disseminated infection. No differences in the protein concentrations of IFN-γ, IL-1β, IL-6, IL-8, IL-10 or TNF-α were observed between isolates of differing genotype. The ability of B. burgdorferi to induce type I and type III IFNs was completely dependent on the presence of linear plasmid (lp) 36. An lp36-deficient B. burgdorferi mutant adhered to, and was internalized by, PBMCs and specific dendritic cell (DC) subsets less efficiently than its isogenic B31 parent strain. The association defect with mDC1s and pDCs could be restored by complementation of the mutant with the complete lp36. The RST1 clinical isolates studied were found to contain a 2.5-kB region, located in the distal one-third of lp36, which was not present in any of the RST3 isolates tested. This divergent region of lp36 may encode one or more factors required for optimal spirochetal recognition and the production of type I and type III IFNs by human DCs, thus suggesting a potential role for DCs in the pathogenesis of B. burgdorferi infection.
Collapse
Affiliation(s)
- Michelle A. Krupna-Gaylord
- Department of Microbiology and Immunology, New York Medical College, Valhalla, New York, United States of America
| | - Dionysios Liveris
- Department of Microbiology and Immunology, New York Medical College, Valhalla, New York, United States of America
| | - Andrea C. Love
- Department of Microbiology and Immunology, New York Medical College, Valhalla, New York, United States of America
| | - Gary P. Wormser
- Division of Infectious Diseases, Department of Medicine, New York Medical College, Valhalla, New York, United States of America
| | - Ira Schwartz
- Department of Microbiology and Immunology, New York Medical College, Valhalla, New York, United States of America
- Division of Infectious Diseases, Department of Medicine, New York Medical College, Valhalla, New York, United States of America
| | - Mary M. Petzke
- Department of Microbiology and Immunology, New York Medical College, Valhalla, New York, United States of America
- * E-mail:
| |
Collapse
|
48
|
Cervantes JL, Hawley KL, Benjamin SJ, Weinerman B, Luu SM, Salazar JC. Phagosomal TLR signaling upon Borrelia burgdorferi infection. Front Cell Infect Microbiol 2014; 4:55. [PMID: 24904837 PMCID: PMC4033037 DOI: 10.3389/fcimb.2014.00055] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 04/09/2014] [Indexed: 12/31/2022] Open
Abstract
Internalization and degradation of live Bb within phagosomal compartments of monocytes, macrophages and dendritic cells (DCs), allows for the release of lipoproteins, nucleic acids and other microbial products, triggering a broad and robust inflammatory response. Toll-like receptors (TLRs) are key players in the recognition of spirochetal ligands from whole viable organisms (i.e., vita-PAMPs). Herein we will review the role of endosomal TLRs in the response to the Lyme disease spirochete.
Collapse
Affiliation(s)
- Jorge L Cervantes
- Department of Pediatrics, University of Connecticut Health Center Farmington, CT, USA ; Division of Infectious Diseases, Connecticut Children's Medical Center Hartford, CT, USA
| | - Kelly L Hawley
- Department of Pediatrics, University of Connecticut Health Center Farmington, CT, USA ; Division of Infectious Diseases, Connecticut Children's Medical Center Hartford, CT, USA
| | - Sarah J Benjamin
- Department of Pediatrics, University of Connecticut Health Center Farmington, CT, USA
| | - Bennett Weinerman
- Department of Pediatrics, University of Connecticut Health Center Farmington, CT, USA
| | - Stephanie M Luu
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center Farmington, CT, USA
| | - Juan C Salazar
- Department of Pediatrics, University of Connecticut Health Center Farmington, CT, USA ; Division of Infectious Diseases, Connecticut Children's Medical Center Hartford, CT, USA ; Department of Immunology, University of Connecticut Health Center Farmington, CT, USA
| |
Collapse
|
49
|
Hoffmann AK, Naj X, Linder S. Daam1 is a regulator of filopodia formation and phagocytic uptake of Borrelia burgdorferi by primary human macrophages. FASEB J 2014; 28:3075-89. [PMID: 24696301 DOI: 10.1096/fj.13-247049] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Borrelia burgdorferi is the causative agent of Lyme disease, an infectious disease that primarily affects the skin, nervous system, and joints. Uptake of borreliae by immune cells is decisive for the course of the infection, and remodelling of the host actin cytoskeleton is crucial in this process. In this study, we showed that the actin-regulatory formin Daam1 is important in Borrelia phagocytosis by primary human macrophages. Uptake of borreliae proceeds preferentially through capture by filopodia and formation of coiling pseudopods that enwrap the spirochetes. Using immunofluorescence, we localized endogenous and overexpressed Daam1 to filopodia and to F-actin-rich uptake structures. Live-cell imaging further showed that Daam1 is enriched at coiling pseudopods that arise from the macrophage surface. This filopodia-independent step was corroborated by control experiments of phagocytic cup formation with latex beads. Moreover, siRNA-mediated knockdown of Daam1 led to a 65% reduction of borreliae-induced filopodia, and, as shown by the outside-inside staining technique, to a 50% decrease in phagocytic uptake of borreliae, as well as a 37% reduction in coiling pseudopod formation. Collectively, we showed that Daam1 plays a dual role in the phagocytic uptake of borreliae: first, as a regulator of filopodia, which are used for capturing spirochetes, and second, in the formation of the coiling pseudopod that enwraps the bacterial cell. These data identify Daam1 as a novel regulator of B. burgdorferi phagocytosis. At the same time, this is the first demonstration of a role for Daam1 in phagocytic processes in general.-Hoffmann, A.-K., Naj, X., Linder, S. Daam1 is a regulator of filopodia formation and phagocytic uptake of Borrelia burgdorferi by primary human macrophages.
Collapse
Affiliation(s)
- Ann-Kathrin Hoffmann
- Institute for Medical Microbiology, Virology, and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| | - Xenia Naj
- Institute for Medical Microbiology, Virology, and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| | - Stefan Linder
- Institute for Medical Microbiology, Virology, and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| |
Collapse
|
50
|
Lovewell RR, Patankar YR, Berwin B. Mechanisms of phagocytosis and host clearance of Pseudomonas aeruginosa. Am J Physiol Lung Cell Mol Physiol 2014; 306:L591-603. [PMID: 24464809 DOI: 10.1152/ajplung.00335.2013] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic bacterial pathogen responsible for a high incidence of acute and chronic pulmonary infection. These infections are particularly prevalent in patients with chronic obstructive pulmonary disease and cystic fibrosis: much of the morbidity and pathophysiology associated with these diseases is due to a hypersusceptibility to bacterial infection. Innate immunity, primarily through inflammatory cytokine production, cellular recruitment, and phagocytic clearance by neutrophils and macrophages, is the key to endogenous control of P. aeruginosa infection. In this review, we highlight recent advances toward understanding the innate immune response to P. aeruginosa, with a focus on the role of phagocytes in control of P. aeruginosa infection. Specifically, we summarize the cellular and molecular mechanisms of phagocytic recognition and uptake of P. aeruginosa, and how current animal models of P. aeruginosa infection reflect clinical observations in the context of phagocytic clearance of the bacteria. Several notable phenotypic changes to the bacteria are consistently observed during chronic pulmonary infections, including changes to mucoidy and flagellar motility, that likely enable or reflect their ability to persist. These traits are likewise examined in the context of how the bacteria avoid phagocytic clearance, inflammation, and sterilizing immunity.
Collapse
Affiliation(s)
- Rustin R Lovewell
- Dept. of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, 1 Medical Center Dr., Lebanon, NH 03756.
| | | | | |
Collapse
|