1
|
Samaddar S, Rolandelli A, O'Neal AJ, Laukaitis-Yousey HJ, Marnin L, Singh N, Wang X, Butler LR, Rangghran P, Kitsou C, Cabrera Paz FE, Valencia L, R Ferraz C, Munderloh UG, Khoo B, Cull B, Rosche KL, Shaw DK, Oliver J, Narasimhan S, Fikrig E, Pal U, Fiskum GM, Polster BM, Pedra JHF. Bacterial reprogramming of tick metabolism impacts vector fitness and susceptibility to infection. Nat Microbiol 2024; 9:2278-2291. [PMID: 38997520 DOI: 10.1038/s41564-024-01756-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/11/2024] [Indexed: 07/14/2024]
Abstract
Arthropod-borne pathogens are responsible for hundreds of millions of infections in humans each year. The blacklegged tick, Ixodes scapularis, is the predominant arthropod vector in the United States and is responsible for transmitting several human pathogens, including the Lyme disease spirochete Borrelia burgdorferi and the obligate intracellular rickettsial bacterium Anaplasma phagocytophilum, which causes human granulocytic anaplasmosis. However, tick metabolic response to microbes and whether metabolite allocation occurs upon infection remain unknown. Here we investigated metabolic reprogramming in the tick ectoparasite I. scapularis and determined that the rickettsial bacterium A. phagocytophilum and the spirochete B. burgdorferi induced glycolysis in tick cells. Surprisingly, the endosymbiont Rickettsia buchneri had a minimal effect on bioenergetics. An unbiased metabolomics approach following A. phagocytophilum infection of tick cells showed alterations in carbohydrate, lipid, nucleotide and protein metabolism, including elevated levels of the pleiotropic metabolite β-aminoisobutyric acid. We manipulated the expression of genes associated with β-aminoisobutyric acid metabolism in I. scapularis, resulting in feeding impairment, diminished survival and reduced bacterial acquisition post haematophagy. Collectively, we discovered that metabolic reprogramming affects interspecies relationships and fitness in the clinically relevant tick I. scapularis.
Collapse
Affiliation(s)
- Sourabh Samaddar
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Agustin Rolandelli
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Anya J O'Neal
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hanna J Laukaitis-Yousey
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Liron Marnin
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Nisha Singh
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Department of Biotechnology, School of Energy Technology, Pandit Deendayal Energy University; Knowledge Corridor, Gandhinagar, India
| | - Xiaowei Wang
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
- MP Biomedicals, Solon, OH, USA
| | - L Rainer Butler
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Parisa Rangghran
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Chrysoula Kitsou
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Francy E Cabrera Paz
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Luisa Valencia
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Camila R Ferraz
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | | | - Benedict Khoo
- Division of Environmental Health Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Benjamin Cull
- Department of Entomology, University of Minnesota, Saint Paul, MN, USA
| | - Kristin L Rosche
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Dana K Shaw
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Jonathan Oliver
- Division of Environmental Health Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Sukanya Narasimhan
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Erol Fikrig
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Gary M Fiskum
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Brian M Polster
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
2
|
Yuan C, Xu Q, Ning Y, Xia Q. Potential mechanisms implied in tick infection by arboviruses and their transmission to vertebrate hosts. Integr Zool 2024. [PMID: 39016029 DOI: 10.1111/1749-4877.12875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Ticks can transmit many pathogens, including arboviruses, to their vertebrate hosts. Arboviruses must overcome or evade defense mechanisms during their passage from the tick gut to the hemolymph, salivary glands, and the feeding site in the host skin. This review summarizes current knowledge of defense mechanisms in specific tick tissues and at the feeding site in the host skin. We discuss the possible roles of these defense mechanisms in viral infection and transmission. The responses of tick salivary proteins to arbovirus infection are also discussed. This review provides information that may help accelerate research on virus-tick interactions.
Collapse
Affiliation(s)
- Chuanfei Yuan
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, China
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Qiong Xu
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, China
| | - Yunjia Ning
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
| | - Qianfeng Xia
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, Hainan Medical University, Haikou, China
| |
Collapse
|
3
|
Gou F, Zhang D, Chen S, Zhang M, Chen J. Role of nuclear protein Akirin in the modulation of female reproduction in Nilaparvata lugens (Hemiptera: Delphacidae). Front Physiol 2024; 15:1415746. [PMID: 39045218 PMCID: PMC11264338 DOI: 10.3389/fphys.2024.1415746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction: Akirin as a highly conserved transcription factor, exerts a profound influence on the growth, development, immune response, and reproductive processes in animals. The brown planthopper (BPH), Nilaparvata lugens, a major pest in rice production in Asia, possesses high reproductive capacity, a critical factor contributing to reduced rice yields. The aims of this study were to demonstrate the regulatory role of Akirin in the reproduction of BPH. Methods: In this study, quantitative PCR (qPCR) was used to detect the mRNA expression of genes. RNA interference (RNAi) was used to downregulate the expression of Akirin gene, and RNA sequencing (RNA-seq) was used to screen for differentially expressed genes caused by Akirin downregulation. Hormone contents were measured with the enzyme linked immunosorbent assay (ELISA), and protein content was evaluated with the bicinchoninic acid (BCA) method. Results: Using BPH genome data, we screened for an Akirin gene (NlAkirin). An analysis of tissue-specific expressions showed that NlAkirin was expressed in all tissues tested in female BPH, but its expression level was highest in the ovary. After inhibiting the mRNA expression of NlAkirin in BPH females, the number of eggs laid, hatching rate, and number of ovarioles decreased. Transcriptome sequencing was performed, following a NlAkirin double-stranded RNA treatment. Compared with the genes of the control, which was injected with GFP double-stranded RNA, there were 438 upregulated genes and 1012 downregulated genes; the expression of vitellogenin (Vg) and vitellogenin receptor (VgR) genes as well as the mRNA expression of genes related to the target of rapamycin (TOR), juvenile hormone (JH), and insulin pathways involved in Vg synthesis was significantly downregulated. As a result of NlAkirin knockdown, the titers of JH III and Ecdysone (Ecd) were downregulated in unmated females but returned to normal levels in mated females. The ovarian protein contents in both unmated and mated females were downregulated. Discussion and conclusion: Our results suggest that NlAkirin affects female BPH reproduction by regulating the mRNA expression of genes related to the Vg, VgR, TOR, JH, and insulin signaling pathways, in addition to the titers of JH III and Ecd. The findings of this research provide novel insights into the regulatory role of Akirin in insect reproductive capacity.
Collapse
Affiliation(s)
- Feiyan Gou
- College of Basic Medical Science, Zunyi Medical University, Zunyi, China
| | - Daowei Zhang
- School of Biological and Agricultural Science and Technology, Zunyi Normal University, Zunyi, China
| | - Siqi Chen
- College of Basic Medical Science, Zunyi Medical University, Zunyi, China
| | - Mingjing Zhang
- College of Basic Medical Science, Zunyi Medical University, Zunyi, China
| | - Jing Chen
- College of Basic Medical Science, Zunyi Medical University, Zunyi, China
| |
Collapse
|
4
|
Feng T, Tong H, Zhang F, Zhang Q, Zhang H, Zhou X, Ruan H, Wu Q, Dai J. Transcriptome study reveals tick immune genes restrict Babesia microti infection. INSECT SCIENCE 2024. [PMID: 38837613 DOI: 10.1111/1744-7917.13384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 06/07/2024]
Abstract
A systems biology approach was employed to gain insight into tick biology and interactions between vectors and pathogens. Haemaphysalis longicornis serves as one of the primary vectors of Babesia microti, significantly impacting human and animal health. Obtaining more information about their relationship is crucial for a comprehensive understanding of tick and pathogen biology, pathogen transmission dynamics, and potential control strategies. RNA sequencing of uninfected and B. microti-infected ticks resulted in the identification of 15 056 unigenes. Among these, 1 051 were found to be differentially expressed, with 796 being upregulated and 255 downregulated (P < 0.05). Integrated transcriptomics datasets revealed the pivotal role of immune-related pathways, including the Toll, Janus kinase/signal transducer and activator of transcription (JAK-STAT), immunodeficiency, and RNA interference (RNAi) pathways, in response to infection. Consequently, 3 genes encoding critical transcriptional factor Dorsal, Relish, and STAT were selected for RNAi experiments. The knockdown of Dorsal, Relish, and STAT resulted in a substantial increase in Babesia infection levels compared to the respective controls. These findings significantly advanced our understanding of tick-Babesia molecular interactions and proposed novel tick antigens as potential vaccine targets against tick infestations and pathogen transmission.
Collapse
Affiliation(s)
- Tingting Feng
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu Province, China
- Central Laboratory, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Tong
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu Province, China
| | - Feihu Zhang
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu Province, China
| | - Qianqian Zhang
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu Province, China
| | - Heng Zhang
- Pasteurien College, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Xia Zhou
- School of Biology and Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Hang Ruan
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu Province, China
| | - Qihan Wu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Jianfeng Dai
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
5
|
Namjoshi P, Lubembe DM, Sultana H, Neelakanta G. Antibody-blocking of a tick transporter impairs Anaplasma phagocytophilum colonization in Haemaphysalis longicornis ticks. Sci Rep 2024; 14:9003. [PMID: 38637614 PMCID: PMC11026487 DOI: 10.1038/s41598-024-59315-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 04/09/2024] [Indexed: 04/20/2024] Open
Abstract
The invasive Asian longhorned tick Haemaphysalis longicornis that vectors and transmits several animal pathogens is significantly expanding in the United States. Recent studies report that these ticks also harbor human pathogens including Borrelia burgdorferi sensu lato, Babesia microti, and Anaplasma phagocytophilum. Therefore, studies that address the interactions of these ticks with human pathogens are important. In this study, we report the characterization of H. longicornis organic anion-transporting polypeptides (OATPs) in interactions of these ticks with A. phagocytophilum. Using OATP-signature sequence, we identified six OATPs in the H. longicornis genome. Bioinformatic analysis revealed that H. longicornis OATPs are closer to other tick orthologs rather than to mammalian counterparts. Quantitative real-time PCR analysis revealed that OATPs are highly expressed in immature stages when compared to mature stages of these ticks. In addition, we noted that the presence of A. phagocytophilum upregulates a specific OATP in these ticks. We also noted that exogenous treatment of H. longicornis with xanthurenic acid, a tryptophan metabolite, influenced OATP expression in these ticks. Immunoblotting analysis revealed that antibody generated against Ixodes scapularis OATP cross-reacted with H. longicornis OATP. Furthermore, treatment of H. longicornis with OATP antibody impaired colonization of A. phagocytophilum in these ticks. These results not only provide evidence that the OATP-tryptophan pathway is important for A. phagocytophilum survival in H. longicornis ticks but also indicate OATP as a promising candidate for the development of a universal anti-tick vaccine to target this bacterium and perhaps other rickettsial pathogens of medical importance.
Collapse
Affiliation(s)
- Prachi Namjoshi
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, 37996, USA
| | - Donald M Lubembe
- Department of Veterinary Pathology, Microbiology and Parasitology, Faculty of Veterinary Medicine and Surgery, Egerton University, Egerton, Kenya
| | - Hameeda Sultana
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, 37996, USA
| | - Girish Neelakanta
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, 37996, USA.
| |
Collapse
|
6
|
Singh N, Rolandelli A, O’Neal AJ, Butler LR, Samaddar S, Laukaitis-Yousey HJ, Butnaru M, Mohr SE, Perrimon N, Pedra JHF. Genetic manipulation of an Ixodes scapularis cell line. mBio 2024; 15:e0247923. [PMID: 38380961 PMCID: PMC10936194 DOI: 10.1128/mbio.02479-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/08/2024] [Indexed: 02/22/2024] Open
Abstract
Although genetic manipulation is one of the hallmarks of model organisms, its applicability to non-model species has remained difficult due to our limited understanding of their fundamental biology. For instance, manipulation of a cell line originated from the black-legged tick Ixodes scapularis, an arthropod that serves as a vector for several human pathogens, has yet to be established. Here, we demonstrate the successful genetic modification of the commonly used tick ISE6 line through ectopic expression and clustered regularly interspaced palindromic repeats [(CRISPR)/CRISPR-associated protein 9 (Cas9)] genome editing. We performed ectopic expression using nucleofection and attained CRISPR-Cas9 editing via homology-dependent recombination. Targeting the E3 ubiquitin ligase x-linked inhibitor of apoptosis (xiap) and its substrate p47 led to an alteration in molecular signaling within the immune deficiency network and increased infection of the rickettsial agent Anaplasma phagocytophilum in I. scapularis ISE6 cells. Collectively, our findings complement techniques for the genetic engineering of I. scapularis ticks, which currently limit efficient and scalable molecular genetic screens in vivo.IMPORTANCEGenetic engineering in arachnids has lagged compared to insects, largely because of substantial differences in their biology. This study unveils the implementation of ectopic expression and CRISPR-Cas9 gene editing in a tick cell line. We introduced fluorescently tagged proteins in ISE6 cells and edited its genome via homology-dependent recombination. We ablated the expression of xiap and p47, two signaling molecules present in the immune deficiency (IMD) pathway of Ixodes scapularis. Impairment of the tick IMD pathway, an analogous network of the tumor necrosis factor receptor in mammals, led to enhanced infection of the rickettsial agent Anaplasma phagocytophilum. Altogether, our findings provide a critical technical resource to the scientific community to enable a deeper understanding of biological circuits in the black-legged tick I. scapularis.
Collapse
Affiliation(s)
- Nisha Singh
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Agustin Rolandelli
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Anya J. O’Neal
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - L. Rainer Butler
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Sourabh Samaddar
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Hanna J. Laukaitis-Yousey
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Matthew Butnaru
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Stephanie E. Mohr
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Joao H. F. Pedra
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Rolandelli A, Laukaitis-Yousey HJ, Bogale HN, Singh N, Samaddar S, O'Neal AJ, Ferraz CR, Butnaru M, Mameli E, Xia B, Mendes MT, Butler LR, Marnin L, Cabrera Paz FE, Valencia LM, Rana VS, Skerry C, Pal U, Mohr SE, Perrimon N, Serre D, Pedra JHF. Tick hemocytes have a pleiotropic role in microbial infection and arthropod fitness. Nat Commun 2024; 15:2117. [PMID: 38459063 PMCID: PMC10923820 DOI: 10.1038/s41467-024-46494-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 02/28/2024] [Indexed: 03/10/2024] Open
Abstract
Uncovering the complexity of systems in non-model organisms is critical for understanding arthropod immunology. Prior efforts have mostly focused on Dipteran insects, which only account for a subset of existing arthropod species in nature. Here we use and develop advanced techniques to describe immune cells (hemocytes) from the clinically relevant tick Ixodes scapularis at a single-cell resolution. We observe molecular alterations in hemocytes upon feeding and infection with either the Lyme disease spirochete Borrelia burgdorferi or the rickettsial agent Anaplasma phagocytophilum. We reveal hemocyte clusters exhibiting defined signatures related to immunity, metabolism, and proliferation. Depletion of phagocytic hemocytes affects hemocytin and astakine levels, two I. scapularis hemocyte markers, impacting blood-feeding, molting behavior, and bacterial acquisition. Mechanistically, astakine alters hemocyte proliferation, whereas hemocytin affects the c-Jun N-terminal kinase (JNK) signaling pathway in I. scapularis. Altogether, we discover a role for tick hemocytes in immunophysiology and provide a valuable resource for comparative biology in arthropods.
Collapse
Affiliation(s)
- Agustin Rolandelli
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hanna J Laukaitis-Yousey
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Haikel N Bogale
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Rancho BioSciences, San Diego, CA, USA
| | - Nisha Singh
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biotechnology, School of Energy Technology, Pandit Deendayal Energy University; Knowledge Corridor, Gandhinagar, Gujarat, India
| | - Sourabh Samaddar
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Anya J O'Neal
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Camila R Ferraz
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Matthew Butnaru
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Enzo Mameli
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, MA, USA
| | - Baolong Xia
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - M Tays Mendes
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - L Rainer Butler
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Liron Marnin
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Francy E Cabrera Paz
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Luisa M Valencia
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Vipin S Rana
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Ciaran Skerry
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Stephanie E Mohr
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - David Serre
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
8
|
Nepveu-Traversy ME, Fausther-Bovendo H, Babuadze G(G. Human Tick-Borne Diseases and Advances in Anti-Tick Vaccine Approaches: A Comprehensive Review. Vaccines (Basel) 2024; 12:141. [PMID: 38400125 PMCID: PMC10891567 DOI: 10.3390/vaccines12020141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
This comprehensive review explores the field of anti-tick vaccines, addressing their significance in combating tick-borne diseases of public health concern. The main objectives are to provide a brief epidemiology of diseases affecting humans and a thorough understanding of tick biology, traditional tick control methods, the development and mechanisms of anti-tick vaccines, their efficacy in field applications, associated challenges, and future prospects. Tick-borne diseases (TBDs) pose a significant and escalating threat to global health and the livestock industries due to the widespread distribution of ticks and the multitude of pathogens they transmit. Traditional tick control methods, such as acaricides and repellents, have limitations, including environmental concerns and the emergence of tick resistance. Anti-tick vaccines offer a promising alternative by targeting specific tick proteins crucial for feeding and pathogen transmission. Developing vaccines with antigens based on these essential proteins is likely to disrupt these processes. Indeed, anti-tick vaccines have shown efficacy in laboratory and field trials successfully implemented in livestock, reducing the prevalence of TBDs. However, some challenges still remain, including vaccine efficacy on different hosts, polymorphisms in ticks of the same species, and the economic considerations of adopting large-scale vaccine strategies. Emerging technologies and approaches hold promise for improving anti-tick vaccine development and expanding their impact on public health and agriculture.
Collapse
Affiliation(s)
| | - Hugues Fausther-Bovendo
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 75550, USA;
| | - George (Giorgi) Babuadze
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 75550, USA;
| |
Collapse
|
9
|
Nandy K, Tamakloe C, Sonenshine DE, Sultana H, Neelakanta G. Anti-tick vaccine candidate subolesin is important for blood feeding and innate immune gene expression in soft ticks. PLoS Negl Trop Dis 2023; 17:e0011719. [PMID: 37934730 PMCID: PMC10629623 DOI: 10.1371/journal.pntd.0011719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 10/11/2023] [Indexed: 11/09/2023] Open
Abstract
Subolesin is a conserved molecule in both hard and soft ticks and is considered as an effective candidate molecule for the development of anti-tick vaccine. Previous studies have reported the role of subolesin in blood feeding, reproduction, development, and gene expression in hard ticks. However, studies addressing the role of subolesin in soft ticks are limited. In this study, we report that subolesin is not only important in soft tick Ornithodoros turicata americanus blood feeding but also in the regulation of innate immune gene expression in these ticks. We identified and characterized several putative innate immune genes including Toll, Lysozyme precursor (Lp), fibrinogen-domain containing protein (FDP), cystatin and ML-domain containing protein (MLD) in O. turicata americanus ticks. Quantitative real-time polymerase chain reaction analysis revealed the expression of these genes in both O. turicata americanus salivary glands and midgut and in all developmental stages of these soft ticks. Significantly increased expression of fdp was noted in salivary glands and midgut upon O. turicata americanus blood feeding. Furthermore, RNAi-mediated knockdown of O. turicata americanus subolesin expression affected blood feeding and innate immune gene expression in these ticks. Significant downregulation of toll, lp, fdp, cystatin, and mld transcripts was evident in sub-dsRNA-treated ticks when compared to the levels noted in mock-dsRNA-treated control. Collectively, our study not only reports identification and characterization of various innate immune genes in O. turicata americanus ticks but also provides evidence on the role of subolesin in blood feeding and innate immune gene expression in these medically important ticks.
Collapse
Affiliation(s)
- Krittika Nandy
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Comfort Tamakloe
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, United States of America
- The University of Queensland- Ochsner Clinical School, Jefferson, Loiusiana, United States of America
| | - Daniel E. Sonenshine
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, United States of America
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Hameeda Sultana
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Girish Neelakanta
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, United States of America
| |
Collapse
|
10
|
Park JM, Genera BM, Fahy D, Swallow KT, Nelson CM, Oliver JD, Shaw DK, Munderloh UG, Brayton KA. An Anaplasma phagocytophilum T4SS effector, AteA, is essential for tick infection. mBio 2023; 14:e0171123. [PMID: 37747883 PMCID: PMC10653876 DOI: 10.1128/mbio.01711-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 07/26/2023] [Indexed: 09/27/2023] Open
Abstract
IMPORTANCE Ticks are the number one vector of pathogens for livestock worldwide and for humans in the United States. The biology of tick transmission is an understudied area. Understanding this critical interaction could provide opportunities to affect the course of disease spread. In this study, we examined the zoonotic tick-borne agent Anaplasma phagocytophilum and identified a secreted protein, AteA, which is expressed in a tick-specific manner. These secreted proteins, termed effectors, are the first proteins to interact with the host environment. AteA is essential for survival in ticks and appears to interact with cortical actin. Most effector proteins are studied in the context of the mammalian host; however, understanding how this unique set of proteins affects tick transmission is critical to developing interventions.
Collapse
Affiliation(s)
- Jason M. Park
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Brittany M. Genera
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Deirdre Fahy
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Kyle T. Swallow
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Curtis M. Nelson
- Department of Entomology, College of Food, Agricultural, and Natural Resource Sciences, University of Minnesota, Saint Paul, Minnesota, USA
| | - Jonathan D. Oliver
- Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Dana K. Shaw
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Ulrike G. Munderloh
- Department of Entomology, College of Food, Agricultural, and Natural Resource Sciences, University of Minnesota, Saint Paul, Minnesota, USA
| | - Kelly A. Brayton
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| |
Collapse
|
11
|
Rosche KL, Hurtado J, Fisk EA, Vosbigian KA, Warren AL, Sidak-Loftis LC, Wright SJ, Ramirez-Zepp E, Park JM, Shaw DK. PERK-mediated antioxidant response is key for pathogen persistence in ticks. mSphere 2023; 8:e0032123. [PMID: 37733353 PMCID: PMC10597351 DOI: 10.1128/msphere.00321-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 07/31/2023] [Indexed: 09/22/2023] Open
Abstract
A crucial phase in the life cycle of tick-borne pathogens is the time spent colonizing and persisting within the arthropod. Tick immunity is emerging as a key force shaping how transmissible pathogens interact with the vector. How pathogens remain in the tick despite immunological pressure remains unknown. In persistently infected Ixodes scapularis, we found that Borrelia burgdorferi (causative agent of Lyme disease) and Anaplasma phagocytophilum (causative agent of granulocytic anaplasmosis) activate a cellular stress pathway mediated by the endoplasmic reticulum receptor PKR-like ER kinase (PERK) and the central regulatory molecule eIF2α. Disabling the PERK pathway through pharmacological inhibition and RNA interference (RNAi) significantly decreased microbial numbers. In vivo RNAi of the PERK pathway not only reduced the number of A. phagocytophilum and B. burgdorferi colonizing larvae after a bloodmeal but also significantly reduced the number of bacteria that survive the molt. An investigation into PERK pathway-regulated targets revealed that A. phagocytophilum and B. burgdorferi induce activity of the antioxidant response regulator, nuclear factor erythroid 2-related factor 2 (Nrf2). Tick cells deficient for nrf2 expression or PERK signaling showed accumulation of reactive oxygen and nitrogen species in addition to reduced microbial survival. Supplementation with antioxidants rescued the microbicidal phenotype caused by blocking the PERK pathway. Altogether, our study demonstrates that the Ixodes PERK pathway is activated by transmissible microbes and facilitates persistence in the arthropod by potentiating an Nrf2-regulated antioxidant environment. IMPORTANCE Recent advances demonstrate that the tick immune system recognizes and limits the pathogens they transmit. Innate immune mediators such as antimicrobial peptides and reactive oxygen/nitrogen species are produced and restrict microbial survival. It is currently unclear how pathogens remain in the tick, despite this immune assault. We found that an antioxidant response controlled by the PERK branch of the unfolded protein response is activated in ticks that are persistently infected with Borrelia burgdorferi (Lyme disease) or Anaplasma phagocytophilum (granulocytic anaplasmosis). The PERK pathway induces the antioxidant response transcription factor, Nrf2, which coordinates a gene network that ultimately neutralizes reactive oxygen and nitrogen species. Interfering with this signaling cascade in ticks causes a significant decline in pathogen numbers. Given that innate immune products can cause collateral damage to host tissues, we speculate that this is an arthropod-driven response aimed at minimizing damage to "self" that also inadvertently benefits the pathogen. Collectively, our findings shed light on the mechanistic push and pull between tick immunity and pathogen persistence within the arthropod vector.
Collapse
Affiliation(s)
- Kristin L. Rosche
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Joanna Hurtado
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Elis A. Fisk
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Kaylee A. Vosbigian
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Ashley L. Warren
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Lindsay C. Sidak-Loftis
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Sarah J. Wright
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Elisabeth Ramirez-Zepp
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Jason M. Park
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Dana K. Shaw
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| |
Collapse
|
12
|
Rolandelli A, Laukaitis-Yousey HJ, Bogale HN, Singh N, Samaddar S, O’Neal AJ, Ferraz CR, Butnaru M, Mameli E, Xia B, Mendes MT, Butler LR, Marnin L, Cabrera Paz FE, Valencia LM, Rana VS, Skerry C, Pal U, Mohr SE, Perrimon N, Serre D, Pedra JH. Tick hemocytes have pleiotropic roles in microbial infection and arthropod fitness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.31.555785. [PMID: 37693411 PMCID: PMC10491215 DOI: 10.1101/2023.08.31.555785] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Uncovering the complexity of systems in non-model organisms is critical for understanding arthropod immunology. Prior efforts have mostly focused on Dipteran insects, which only account for a subset of existing arthropod species in nature. Here, we describe immune cells or hemocytes from the clinically relevant tick Ixodes scapularis using bulk and single cell RNA sequencing combined with depletion via clodronate liposomes, RNA interference, Clustered Regularly Interspaced Short Palindromic Repeats activation (CRISPRa) and RNA-fluorescence in situ hybridization (FISH). We observe molecular alterations in hemocytes upon tick infestation of mammals and infection with either the Lyme disease spirochete Borrelia burgdorferi or the rickettsial agent Anaplasma phagocytophilum. We predict distinct hemocyte lineages and reveal clusters exhibiting defined signatures for immunity, metabolism, and proliferation during hematophagy. Furthermore, we perform a mechanistic characterization of two I. scapularis hemocyte markers: hemocytin and astakine. Depletion of phagocytic hemocytes affects hemocytin and astakine levels, which impacts blood feeding and molting behavior of ticks. Hemocytin specifically affects the c-Jun N-terminal kinase (JNK) signaling pathway, whereas astakine alters hemocyte proliferation in I. scapularis. Altogether, we uncover the heterogeneity and pleiotropic roles of hemocytes in ticks and provide a valuable resource for comparative biology in arthropods.
Collapse
Affiliation(s)
- Agustin Rolandelli
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Hanna J. Laukaitis-Yousey
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Haikel N. Bogale
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Nisha Singh
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sourabh Samaddar
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Anya J. O’Neal
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Camila R. Ferraz
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Matthew Butnaru
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Enzo Mameli
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Baolong Xia
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - M. Tays Mendes
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - L. Rainer Butler
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Liron Marnin
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Francy E. Cabrera Paz
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Luisa M. Valencia
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Vipin S. Rana
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Ciaran Skerry
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Stephanie E. Mohr
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - David Serre
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Joao H.F. Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
13
|
Shi XZ, Yang MC, Kang XL, Li YX, Hong PP, Zhao XF, Vasta G, Wang JX. Scavenger receptor B2, a type III membrane pattern recognition receptor, senses LPS and activates the IMD pathway in crustaceans. Proc Natl Acad Sci U S A 2023; 120:e2216574120. [PMID: 37276415 PMCID: PMC10268257 DOI: 10.1073/pnas.2216574120] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 04/24/2023] [Indexed: 06/07/2023] Open
Abstract
The immune deficiency (IMD) pathway is critical for elevating host immunity in both insects and crustaceans. The IMD pathway activation in insects is mediated by peptidoglycan recognition proteins, which do not exist in crustaceans, suggesting a previously unidentified mechanism involved in crustacean IMD pathway activation. In this study, we identified a Marsupenaeus japonicus B class type III scavenger receptor, SRB2, as a receptor for activation of the IMD pathway. SRB2 is up-regulated upon bacterial challenge, while its depletion exacerbates bacterial proliferation and shrimp mortality via abolishing the expression of antimicrobial peptides. The extracellular domain of SRB2 recognizes bacterial lipopolysaccharide (LPS), while its C-terminal intracellular region containing a cryptic RHIM-like motif interacts with IMD, and activates the pathway by promoting nuclear translocation of RELISH. Overexpressing shrimp SRB2 in Drosophila melanogaster S2 cells potentiates LPS-induced IMD pathway activation and diptericin expression. These results unveil a previously unrecognized SRB2-IMD axis responsible for antimicrobial peptide induction and restriction of bacterial infection in crustaceans and provide evidence of biological diversity of IMD signaling in animals. A better understanding of the innate immunity of crustaceans will permit the optimization of prevention and treatment strategies against the arising shrimp diseases.
Collapse
Affiliation(s)
- Xiu-Zhen Shi
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, 266237, Qingdao, Shandong, China
| | - Ming-Chong Yang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, 266237, Qingdao, Shandong, China
- State Key Laboratory of Microbial Technology, Shandong University, 266237, Qingdao, Shandong, China
| | - Xin-Le Kang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, 266237, Qingdao, Shandong, China
| | - Yan-Xue Li
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, 266237, Qingdao, Shandong, China
| | - Pan-Pan Hong
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, 266237, Qingdao, Shandong, China
| | - Xiao-Fan Zhao
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, 266237, Qingdao, Shandong, China
| | - Gerardo R. Vasta
- Department of Microbiology and Immunology, School of Medicine, Institute of Marine and Environmental Technology, University of Maryland Baltimore, Baltimore, MD21202
| | - Jin-Xing Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, 266237, Qingdao, Shandong, China
- State Key Laboratory of Microbial Technology, Shandong University, 266237, Qingdao, Shandong, China
| |
Collapse
|
14
|
Rosche KL, Hurtado J, Fisk EA, Vosbigian KA, Warren AL, Sidak-Loftis LC, Wright SJ, Ramirez-Zepp E, Park JM, Shaw DK. PERK-mediated antioxidant response is key for pathogen persistence in ticks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542958. [PMID: 37398437 PMCID: PMC10312570 DOI: 10.1101/2023.05.30.542958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
A crucial phase in the lifecycle of tick-borne pathogens is the time spent colonizing and persisting within the arthropod. Tick immunity is emerging as a key force shaping how transmissible pathogens interact with the vector. How pathogens remain in the tick despite immunological pressure remains unknown. In persistently infected Ixodes scapularis , we found that Borrelia burgdorferi (Lyme disease) and Anaplasma phagocytophilum (granulocytic anaplasmosis) activate a cellular stress pathway mediated by the endoplasmic reticulum receptor PERK and the central regulatory molecule, eIF2α. Disabling the PERK pathway through pharmacological inhibition and RNAi significantly decreased microbial numbers. In vivo RNA interference of the PERK pathway not only reduced the number of A. phagocytophilum and B. burgdorferi colonizing larvae after a bloodmeal, but also significantly reduced the number of bacteria that survive the molt. An investigation into PERK pathway-regulated targets revealed that A. phagocytophilum and B. burgdorferi induce activity of the antioxidant response regulator, Nrf2. Tick cells deficient for nrf2 expression or PERK signaling showed accumulation of reactive oxygen and nitrogen species in addition to reduced microbial survival. Supplementation with antioxidants rescued the microbicidal phenotype caused by blocking the PERK pathway. Altogether, our study demonstrates that the Ixodes PERK pathway is activated by transmissible microbes and facilitates persistence in the arthropod by potentiating an Nrf2-regulated antioxidant environment.
Collapse
Affiliation(s)
- Kristin L. Rosche
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Joanna Hurtado
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Elis A. Fisk
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Kaylee A. Vosbigian
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Ashley L. Warren
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Lindsay C. Sidak-Loftis
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Sarah J. Wright
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Elisabeth Ramirez-Zepp
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Jason M. Park
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Dana K. Shaw
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| |
Collapse
|
15
|
Samaddar S, O'Neal AJ, Marnin L, Rolandelli A, Singh N, Wang X, Butler LR, Rangghran P, Laukaitis HJ, Cabrera Paz FE, Fiskum GM, Polster BM, Pedra JHF. Metabolic disruption impacts tick fitness and microbial relationships. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.26.542501. [PMID: 37292783 PMCID: PMC10245996 DOI: 10.1101/2023.05.26.542501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Arthropod-borne microbes rely on the metabolic state of a host to cycle between evolutionarily distant species. For instance, arthropod tolerance to infection may be due to redistribution of metabolic resources, often leading to microbial transmission to mammals. Conversely, metabolic alterations aids in pathogen elimination in humans, who do not ordinarily harbor arthropod-borne microbes. To ascertain the effect of metabolism on interspecies relationships, we engineered a system to evaluate glycolysis and oxidative phosphorylation in the tick Ixodes scapularis. Using a metabolic flux assay, we determined that the rickettsial bacterium Anaplasma phagocytophilum and the Lyme disease spirochete Borrelia burgdorferi, which are transstadially transmitted in nature, induced glycolysis in ticks. On the other hand, the endosymbiont Rickettsia buchneri, which is transovarially maintained, had a minimal effect on I. scapularis bioenergetics. Importantly, the metabolite β-aminoisobutyric acid (BAIBA) was elevated during A. phagocytophilum infection of tick cells following an unbiased metabolomics approach. Thus, we manipulated the expression of genes associated with the catabolism and anabolism of BAIBA in I. scapularis and detected impaired feeding on mammals, reduced bacterial acquisition, and decreased tick survival. Collectively, we reveal the importance of metabolism for tick-microbe relationships and unveil a valuable metabolite for I. scapularis fitness.
Collapse
Affiliation(s)
- Sourabh Samaddar
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - Anya J O'Neal
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - Liron Marnin
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - Agustin Rolandelli
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - Nisha Singh
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - Xiaowei Wang
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - L Rainer Butler
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - Parisa Rangghran
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Hanna J Laukaitis
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - Francy E Cabrera Paz
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - Gary M Fiskum
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Brian M Polster
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| |
Collapse
|
16
|
Mazuecos L, Alberdi P, Hernández-Jarguín A, Contreras M, Villar M, Cabezas-Cruz A, Simo L, González-García A, Díaz-Sánchez S, Neelakanta G, Bonnet SI, Fikrig E, de la Fuente J. Frankenbacteriosis targeting interactions between pathogen and symbiont to control infection in the tick vector. iScience 2023; 26:106697. [PMID: 37168564 PMCID: PMC10165458 DOI: 10.1016/j.isci.2023.106697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/23/2023] [Accepted: 04/13/2023] [Indexed: 05/13/2023] Open
Abstract
Tick microbiota can be targeted for the control of tick-borne diseases such as human granulocytic anaplasmosis (HGA) caused by model pathogen, Anaplasma phagocytophilum. Frankenbacteriosis is inspired by Frankenstein and defined here as paratransgenesis of tick symbiotic/commensal bacteria to mimic and compete with tick-borne pathogens. Interactions between A. phagocytophilum and symbiotic Sphingomonas identified by metaproteomics analysis in Ixodes scapularis midgut showed competition between both bacteria. Consequently, Sphingomonas was selected for frankenbacteriosis for the control of A. phagocytophilum infection and transmission. The results showed that Franken Sphingomonas producing A. phagocytophilum major surface protein 4 (MSP4) mimic pathogen and reduce infection in ticks by competition and interaction with cell receptor components of infection. Franken Sphingomonas-MSP4 transovarial and trans-stadial transmission suggests that tick larvae with genetically modified Franken Sphingomonas-MSP4 could be produced in the laboratory and released in the field to compete and replace the wildtype populations with associated reduction in pathogen infection/transmission and HGA disease risks.
Collapse
Affiliation(s)
- Lorena Mazuecos
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Pilar Alberdi
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Angélica Hernández-Jarguín
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Marinela Contreras
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Margarita Villar
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Alejandro Cabezas-Cruz
- ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700 Maisons-Alfort, France
| | - Ladislav Simo
- ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700 Maisons-Alfort, France
| | - Almudena González-García
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Sandra Díaz-Sánchez
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Girish Neelakanta
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Sarah I. Bonnet
- Functional Genetics of Infectious Diseases Unit, Institut Pasteur, CNRS UMR 2000, Université de Paris, 75015 Paris, France
- Animal Health Department, INRAE, 37380 Nouzilly, France
| | - Erol Fikrig
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 208022, USA
| | - José de la Fuente
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA
- Corresponding author
| |
Collapse
|
17
|
O’Neal A, Singh N, Rolandelli A, Laukaitis HJ, Wang X, Shaw D, Young B, Narasimhan S, Dutta S, Snyder G, Samaddar S, Marnin L, Butler L, Mendes M, Cabrera Paz F, Valencia L, Sundberg E, Fikrig E, Pal U, Weber D, Pedra J. Croquemort elicits activation of the immune deficiency pathway in ticks. Proc Natl Acad Sci U S A 2023; 120:e2208673120. [PMID: 37155900 PMCID: PMC10193931 DOI: 10.1073/pnas.2208673120] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 03/27/2023] [Indexed: 05/10/2023] Open
Abstract
The immune deficiency (IMD) pathway directs host defense in arthropods upon bacterial infection. In Pancrustacea, peptidoglycan recognition proteins sense microbial moieties and initiate nuclear factor-κB-driven immune responses. Proteins that elicit the IMD pathway in non-insect arthropods remain elusive. Here, we show that an Ixodes scapularis homolog of croquemort (Crq), a CD36-like protein, promotes activation of the tick IMD pathway. Crq exhibits plasma membrane localization and binds the lipid agonist 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoglycerol. Crq regulates the IMD and jun N-terminal kinase signaling cascades and limits the acquisition of the Lyme disease spirochete B. burgdorferi. Additionally, nymphs silenced for crq display impaired feeding and delayed molting to adulthood due to a deficiency in ecdysteroid synthesis. Collectively, we establish a distinct mechanism for arthropod immunity outside of insects and crustaceans.
Collapse
Affiliation(s)
- Anya J. O’Neal
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD21201
| | - Nisha Singh
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD21201
| | - Agustin Rolandelli
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD21201
| | - Hanna J. Laukaitis
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD21201
| | - Xiaowei Wang
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD21201
| | - Dana K. Shaw
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD21201
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA99164
| | - Brianna D. Young
- Department of Biochemistry and Molecular Biology, Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD21201
| | - Sukanya Narasimhan
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT06510
| | - Shraboni Dutta
- Department of Veterinary Medicine, University of Maryland, College Park, MD20742
| | - Greg A. Snyder
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD21201
| | - Sourabh Samaddar
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD21201
| | - Liron Marnin
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD21201
| | - L. Rainer Butler
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD21201
| | - M. Tays Mendes
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD21201
| | - Francy E. Cabrera Paz
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD21201
| | - Luisa M. Valencia
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD21201
| | - Eric J. Sundberg
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD21201
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD21201
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA30322
| | - Erol Fikrig
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT06510
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD20742
| | - David J. Weber
- Department of Biochemistry and Molecular Biology, Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Baltimore, MD21201
| | - Joao H. F. Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD21201
| |
Collapse
|
18
|
Adegoke A, Ribeiro JMC, Brown S, Smith RC, Karim S. Rickettsia parkeri hijacks tick hemocytes to manipulate cellular and humoral transcriptional responses. Front Immunol 2023; 14:1094326. [PMID: 36845157 PMCID: PMC9950277 DOI: 10.3389/fimmu.2023.1094326] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/16/2023] [Indexed: 02/12/2023] Open
Abstract
Introduction Blood-feeding arthropods rely on robust cellular and humoral immunity to control pathogen invasion and replication. Tick hemocytes produce factors that can facilitate or suppress microbial infection and pathogenesis. Despite the importance of hemocytes in regulating microbial infection, understanding of their basic biology and molecular mechanisms remains limited. Methods Here we combined histomorphology and functional analysis to identify five distinct phagocytic and non-phagocytic hemocyte populations circulating within the Gulf Coast tick Amblyomma maculatum. Results and discussion Depletion of phagocytic hemocytes using clodronate liposomes revealed their function in eliminating bacterial infection. We provide the first direct evidence that an intracellular tick-borne pathogen, Rickettsia parkeri, infects phagocytic hemocytes in Am. maculatum to modify tick cellular immune responses. A hemocyte-specific RNA-seq dataset generated from hemocytes isolated from uninfected and R. parkeri-infected partially blood-fed ticks generated ~40,000 differentially regulated transcripts, >11,000 of which were immune genes. Silencing two differentially regulated phagocytic immune marker genes (nimrod B2 and eater-two Drosophila homologs), significantly reduced hemocyte phagocytosis. Conclusion Together, these findings represent a significant step forward in understanding how hemocytes regulate microbial homeostasis and vector competence.
Collapse
Affiliation(s)
- Abdulsalam Adegoke
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Jose M. C. Ribeiro
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Sidney Brown
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Ryan C. Smith
- Department of Plant Pathology, Entomology, and Microbiology, Iowa State University, Ames, IA, United States
| | - Shahid Karim
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| |
Collapse
|
19
|
Park JM, Genera BM, Fahy D, Swallow KT, Nelson CM, Oliver JD, Shaw DK, Munderloh UG, Brayton KA. An Anaplasma phagocytophilum T4SS effector, AteA, is essential for tick infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.06.527355. [PMID: 36798287 PMCID: PMC9934581 DOI: 10.1101/2023.02.06.527355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Pathogens must adapt to disparate environments in permissive host species, a feat that is especially pronounced for vector-borne microbes, which transition between vertebrate hosts and arthropod vectors to complete their lifecycles. Most knowledge about arthropod-vectored bacterial pathogens centers on their life in the mammalian host, where disease occurs. However, disease outbreaks are driven by the arthropod vectors. Adapting to the arthropod is critical for obligate intracellular rickettsial pathogens, as they depend on eukaryotic cells for survival. To manipulate the intracellular environment, these bacteria use Type IV Secretion Systems (T4SS) to deliver effectors into the host cell. To date, few rickettsial T4SS translocated effectors have been identified and have only been examined in the context of mammalian infection. We identified an effector from the tick-borne rickettsial pathogen Anaplasma phagocytophilum , HGE1_02492, as critical for survival in tick cells and acquisition by ticks in vivo . Conversely, HGE1_02492 was dispensable during mammalian cell culture and murine infection. We show HGE1_02492 is translocatable in a T4SS-dependent manner to the host cell cytosol. In eukaryotic cells, the HGE1_02492 localized with cortical actin filaments, which is dependent on multiple sub-domains of the protein. HGE1_02492 is the first arthropod-vector specific T4SS translocated effector identified from a rickettsial pathogen. Moreover, the subcellular target of HGE1_02492 suggests that A. phagocytophilum is manipulating actin to enable arthropod colonization. Based on these findings, we propose the name AteA for Anaplasma ( phagocytophilum ) tick effector A. Altogether, we show that A. phagocytophilum uses distinct strategies to cycle between mammals and arthropods. Importance Ticks are the number one vector of pathogens for livestock worldwide and for humans in the US. The biology of tick transmission is an understudied area. Understanding this critical interaction could provide opportunities to affect the course of disease spread. In this study we examined the zoonotic tick-borne agent Anaplasma phagocytophilum and identified a secreted protein, AteA, that is expressed in a tick-specific manner. These secreted proteins, termed effectors, are the first proteins to interact with the host environment. AteA is essential for survival in ticks and appears to interact with cortical actin. Most effector proteins are studied in the context of the mammalian host; however, understanding how this unique set of proteins affect tick transmission is critical to developing interventions.
Collapse
|
20
|
Perveen N, Muhammad K, Muzaffar SB, Zaheer T, Munawar N, Gajic B, Sparagano OA, Kishore U, Willingham AL. Host-pathogen interaction in arthropod vectors: Lessons from viral infections. Front Immunol 2023; 14:1061899. [PMID: 36817439 PMCID: PMC9929866 DOI: 10.3389/fimmu.2023.1061899] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
Haematophagous arthropods can harbor various pathogens including viruses, bacteria, protozoa, and nematodes. Insects possess an innate immune system comprising of both cellular and humoral components to fight against various infections. Haemocytes, the cellular components of haemolymph, are central to the insect immune system as their primary functions include phagocytosis, encapsulation, coagulation, detoxification, and storage and distribution of nutritive materials. Plasmatocytes and granulocytes are also involved in cellular defense responses. Blood-feeding arthropods, such as mosquitoes and ticks, can harbour a variety of viral pathogens that can cause infectious diseases in both human and animal hosts. Therefore, it is imperative to study the virus-vector-host relationships since arthropod vectors are important constituents of the ecosystem. Regardless of the complex immune response of these arthropod vectors, the viruses usually manage to survive and are transmitted to the eventual host. A multidisciplinary approach utilizing novel and strategic interventions is required to control ectoparasite infestations and block vector-borne transmission of viral pathogens to humans and animals. In this review, we discuss the arthropod immune response to viral infections with a primary focus on the innate immune responses of ticks and mosquitoes. We aim to summarize critically the vector immune system and their infection transmission strategies to mammalian hosts to foster debate that could help in developing new therapeutic strategies to protect human and animal hosts against arthropod-borne viral infections.
Collapse
Affiliation(s)
- Nighat Perveen
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Khalid Muhammad
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Sabir Bin Muzaffar
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Tean Zaheer
- Department of Parasitology, University of Agriculture, Faisalabad, Pakistan
| | - Nayla Munawar
- Department of Chemistry, College of Science, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Bojan Gajic
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Olivier Andre Sparagano
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Uday Kishore
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Arve Lee Willingham
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al-Ain, United Arab Emirates
| |
Collapse
|
21
|
Rana VS, Kitsou C, Dutta S, Ronzetti MH, Zhang M, Bernard Q, Smith AA, Tomás-Cortázar J, Yang X, Wu MJ, Kepple O, Li W, Dwyer JE, Matias J, Baljinnyam B, Oliver JD, Rajeevan N, Pedra JHF, Narasimhan S, Wang Y, Munderloh U, Fikrig E, Simeonov A, Anguita J, Pal U. Dome1-JAK-STAT signaling between parasite and host integrates vector immunity and development. Science 2023; 379:eabl3837. [PMID: 36634189 PMCID: PMC10122270 DOI: 10.1126/science.abl3837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 12/08/2022] [Indexed: 01/14/2023]
Abstract
Ancestral signaling pathways serve critical roles in metazoan development, physiology, and immunity. We report an evolutionary interspecies communication pathway involving a central Ixodes scapularis tick receptor termed Dome1, which acquired a mammalian cytokine receptor motif exhibiting high affinity for interferon-gamma (IFN-γ). Host-derived IFN-γ facilitates Dome1-mediated activation of the Ixodes JAK-STAT pathway. This accelerates tick blood meal acquisition and development while upregulating antimicrobial components. The Dome1-JAK-STAT pathway, which exists in most Ixodid tick genomes, regulates the regeneration and proliferation of gut cells-including stem cells-and dictates metamorphosis through the Hedgehog and Notch-Delta networks, ultimately affecting Ixodes vectorial competence. We highlight the evolutionary dependence of I. scapularis on mammalian hosts through cross-species signaling mechanisms that dually influence arthropod immunity and development.
Collapse
Affiliation(s)
- Vipin S. Rana
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Chrysoula Kitsou
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Shraboni Dutta
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Michael H. Ronzetti
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Min Zhang
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Quentin Bernard
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Alexis A. Smith
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Julen Tomás-Cortázar
- CIC bioGUNE-BRTA (Basque Research & Technology Alliance), 48160 Derio, Bizkaia, Spain
| | - Xiuli Yang
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Ming-Jie Wu
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Oleksandra Kepple
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Weizhong Li
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Jennifer E. Dwyer
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jaqueline Matias
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Bolormaa Baljinnyam
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | | | - Nallakkandi Rajeevan
- Yale Center for Medical Informatics, Yale University School of Medicine, New Haven, CT, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sukanya Narasimhan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Yan Wang
- Mass Spectrometry Facility, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Ulrike Munderloh
- Department of Entomology, University of Minnesota, Minneapolis, MN, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Juan Anguita
- CIC bioGUNE-BRTA (Basque Research & Technology Alliance), 48160 Derio, Bizkaia, Spain
- Ikerbasque, Basque Foundation for Science, 48011 Bilbao, Bizkaia, Spain
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
- Virginia-Maryland College of Veterinary Medicine, College Park, MD, USA
| |
Collapse
|
22
|
Rana VS, Kitsou C, Dumler JS, Pal U. Immune evasion strategies of major tick-transmitted bacterial pathogens. Trends Microbiol 2023; 31:62-75. [PMID: 36055896 PMCID: PMC9772108 DOI: 10.1016/j.tim.2022.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/28/2022] [Accepted: 08/01/2022] [Indexed: 12/30/2022]
Abstract
Tick-transmitted bacterial pathogens thrive in enzootic infection cycles, colonizing disparate vertebrate and arthropod tissues, often establishing persistent infections. Therefore, the evolution of robust immune evasion strategies is central to their successful persistence or transmission between hosts. To survive in nature, these pathogens must counteract a broad range of microbicidal host responses that can be localized, tissue-specific, or systemic, including a mix of these responses at the host-vector interface. Herein, we review microbial immune evasion strategies focusing on Lyme disease spirochetes and rickettsial or tularemia agents as models for extracellular and intracellular tick-borne pathogens, respectively. A better understanding of these adaptive strategies could enrich our knowledge of the infection biology of relevant tick-borne diseases, contributing to the development of future preventions.
Collapse
Affiliation(s)
- Vipin Singh Rana
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Chrysoula Kitsou
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - J Stephen Dumler
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA.
| |
Collapse
|
23
|
Differential Expression of Immune Genes in the Rhipicephalus microplus Gut in Response to Theileria equi Infection. Pathogens 2022; 11:pathogens11121478. [PMID: 36558812 PMCID: PMC9782190 DOI: 10.3390/pathogens11121478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Rhipicephalus microplus is the only tick species known to serve as a biological vector of Theileria equi for horses and other equids in Brazil. The protozoan T. equi is one of the causal agents of equine piroplasmosis, a major threat in horse breeding systems. Vector competence is closely linked to the pathogens' ability to evade tick defense mechanisms. However, knowledge of tick immune response against infections by hemoparasites of the Theileria genus is scarce. In the present study, the expression of genes involved in immune signaling pathways of R. microplus adults' guts when challenged with a high or low parasitic load of T. equi was evaluated. This research demonstrates divergences in the immune gene expression pattern linked to T. equi infection in R. microplus since the Toll, IMD, and JNK signaling pathways were transcriptionally repressed in the guts of adult ticks infected with T. equi. Moreover, the results showed that different infectious doses of T. equi induce differential gene expression of key components of immune signaling cascades in R. microplus gut, suggesting a link between the intensity of infection and the activation of tick immunity response. The present study adds knowledge to elucidate the gut immune signaling response of R. microplus to T. equi infection. In addition, the generated data can serve as a basis for further investigations to develop strategies for controlling and preventing equine piroplasmosis.
Collapse
|
24
|
Rickettsial pathogen inhibits tick cell death through tryptophan metabolite mediated activation of p38 MAP kinase. iScience 2022; 26:105730. [PMID: 36582833 PMCID: PMC9792911 DOI: 10.1016/j.isci.2022.105730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/27/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Anaplasma phagocytophilum modulates various cell signaling pathways in mammalian cells for its survival. In this study, we report that A. phagocytophilum modulates tick tryptophan pathway to activate arthropod p38 MAP kinase for the survival of both this bacterium and its vector host. Increased level of tryptophan metabolite, xanthurenic acid (XA), was evident in A. phagocytophilum-infected ticks and tick cells. Lower levels of cell death markers and increased levels of total and phosphorylated p38 MAPK was noted in A. phagocytophilum-infected ticks and tick cells. Treatment with XA increased phosphorylated p38 MAPK levels and reduced cell death in A. phagocytophilum-infected tick cells. Furthermore, treatment with p38 MAPK inhibitor affected bacterial replication, decreased phosphorylated p38 MAPK levels and increased tick cell death. However, XA reversed these effects. Taken together, we provide evidence that rickettsial pathogen modulates arthropod tryptophan and p38 MAPK pathways to inhibit cell death for its survival in ticks.
Collapse
|
25
|
Ma L, Liu S, Lu P, Yan X, Hao C, Wang H, Wei J, Qie X, Lu Z. The IMD pathway in Hemipteran: A comparative analysis and discussion. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 136:104513. [PMID: 35977558 DOI: 10.1016/j.dci.2022.104513] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 06/15/2023]
Abstract
The evolutionary patterns of the genes in the IMD pathway in hemipterans were characterized and compared. The hemipteran insects were clustered into two groups. One group that encompasses whitefly, plant lice, and scale insect partially lacks the IMD pathway and all antimicrobial peptide (AMP) genes, with the vast majority of IMD pathway and all AMP genes being absent in aphids. The reasons for the absence of the IMD pathway and AMP genes in hemipterans were analyzed based on aphids, in terms of fitness costs. In case of limited resources, aphids have to make a trade-off between the necessary costs such as clean food sources, the essential amino acids supplied by primary bacterial symbionts for survival, nutrients and/or protections against stress provided by secondary symbionts, and the high reproductive capacity, and the costs that do not increase the fitness. Obviously, aphids have to abandon the strong immune system, especially the AMPs and IMD pathway which is mainly against Gram-negative bacteria. The common ground shared with aphids may be the reason for the absence of the IMD pathway and AMP genes in other hemipteran insects.
Collapse
Affiliation(s)
- Li Ma
- Department of Plant Protection, College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China; Shanxi Key Laboratory of Integrated Pest Management in Agriculture, Shanxi Agricultural University, Taiyuan, Shanxi, 030801, China; Department of Entomology, College of Plant Protection, Northwest A & F University, Yangling, Shaanxi, 712100, China
| | - Shanlin Liu
- Department of Entomology, College of Plant Protection, China Agricultural University, Beijing, 100083, China
| | - Ping Lu
- Department of Entomology, College of Plant Protection, Northwest A & F University, Yangling, Shaanxi, 712100, China
| | - Xizhong Yan
- Department of Plant Protection, College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Chi Hao
- Department of Plant Protection, College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Han Wang
- Department of Plant Protection, College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Jiufeng Wei
- Department of Plant Protection, College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Xingtao Qie
- Department of Plant Protection, College of Plant Protection, Shanxi Agricultural University, Taigu, Shanxi, 030801, China.
| | - Zhiqiang Lu
- Department of Entomology, College of Plant Protection, Northwest A & F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
26
|
Narasimhan S, Rajeevan N, Graham M, Wu MJ, DePonte K, Marion S, Masson O, O'Neal AJ, Pedra JHF, Sonenshine DE, Fikrig E. Tick transmission of Borrelia burgdorferi to the murine host is not influenced by environmentally acquired midgut microbiota. MICROBIOME 2022; 10:173. [PMID: 36253842 PMCID: PMC9575305 DOI: 10.1186/s40168-022-01378-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 09/20/2022] [Indexed: 06/09/2023]
Abstract
BACKGROUND Ixodes scapularis is the predominant tick vector of Borrelia burgdorferi, the agent of Lyme disease, in the USA. Molecular interactions between the tick and B. burgdorferi orchestrate the migration of spirochetes from the midgut to the salivary glands-critical steps that precede transmission to the vertebrate host. Over the last decade, research efforts have invoked a potential role for the tick microbiome in modulating tick-pathogen interactions. RESULTS Using multiple strategies to perturb the microbiome composition of B. burgdorferi-infected nymphal ticks, we observe that changes in the microbiome composition do not significantly influence B. burgdorferi migration from the midgut, invasion of salivary glands, or transmission to the murine host. We also show that within 24 and 48 h of the onset of tick feeding, B. burgdorferi spirochetes are within the peritrophic matrix and epithelial cells of the midgut in preparation for exit from the midgut. CONCLUSIONS This study highlights two aspects of tick-spirochete interactions: (1) environmental bacteria associated with the tick do not influence spirochete transmission to the mammalian host and (2) the spirochete may utilize an intracellular exit route during migration from the midgut to the salivary glands, a strategy that may allow the spirochete to distance itself from microbiota in the midgut lumen effectively. This may explain in part, the inability of environment-acquired midgut microbiota to significantly influence spirochete transmission. Unraveling a molecular understanding of this exit strategy will be critical to gain new insights into the biology of the spirochete and the tick. Video Abstract.
Collapse
Affiliation(s)
- Sukanya Narasimhan
- Department of Internal Medicine, Section of Infectious Diseases, New Haven, USA.
| | | | - Morven Graham
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06420, USA
| | - Ming-Jie Wu
- Department of Internal Medicine, Section of Infectious Diseases, New Haven, USA
| | - Kathleen DePonte
- Department of Internal Medicine, Section of Infectious Diseases, New Haven, USA
| | - Solenne Marion
- Department of Internal Medicine, Section of Infectious Diseases, New Haven, USA
- Current address: Roche Diagnostics International, 6343, Rotkreuz, Switzerland
| | - Orlanne Masson
- Department of Internal Medicine, Section of Infectious Diseases, New Haven, USA
| | - Anya J O'Neal
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Daniel E Sonenshine
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD, 20852, USA
| | - Erol Fikrig
- Department of Internal Medicine, Section of Infectious Diseases, New Haven, USA
| |
Collapse
|
27
|
Sidak-Loftis LC, Rosche KL, Pence N, Ujczo JK, Hurtado J, Fisk EA, Goodman AG, Noh SM, Peters JW, Shaw DK. The Unfolded-Protein Response Triggers the Arthropod Immune Deficiency Pathway. mBio 2022; 13:e0070322. [PMID: 35862781 PMCID: PMC9426425 DOI: 10.1128/mbio.00703-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/27/2022] [Indexed: 11/20/2022] Open
Abstract
The insect immune deficiency (IMD) pathway is a defense mechanism that senses and responds to Gram-negative bacteria. Ticks lack genes encoding upstream components that initiate the IMD pathway. Despite this deficiency, core signaling molecules are present and functionally restrict tick-borne pathogens. The molecular events preceding activation remain undefined. Here, we show that the unfolded-protein response (UPR) initiates the IMD network. The endoplasmic reticulum (ER) stress receptor IRE1α is phosphorylated in response to tick-borne bacteria but does not splice the mRNA encoding XBP1. Instead, through protein modeling and reciprocal pulldowns, we show that Ixodes IRE1α complexes with TRAF2. Disrupting IRE1α-TRAF2 signaling blocks IMD pathway activation and diminishes the production of reactive oxygen species. Through in vitro, in vivo, and ex vivo techniques, we demonstrate that the UPR-IMD pathway circuitry limits the Lyme disease-causing spirochete Borrelia burgdorferi and the rickettsial agents Anaplasma phagocytophilum and A. marginale (anaplasmosis). Altogether, our study uncovers a novel linkage between the UPR and the IMD pathway in arthropods. IMPORTANCE The ability of an arthropod to harbor and transmit pathogens is termed "vector competency." Many factors influence vector competency, including how arthropod immune processes respond to the microbe. Divergences in innate immunity between arthropods are increasingly being reported. For instance, although ticks lack genes encoding key upstream molecules of the immune deficiency (IMD) pathway, it is still functional and restricts causative agents of Lyme disease (Borrelia burgdorferi) and anaplasmosis (Anaplasma phagocytophilum). How the IMD pathway is activated in ticks without classically defined pathway initiators is not known. Here, we found that a cellular stress response network, the unfolded-protein response (UPR), functions upstream to induce the IMD pathway and restrict transmissible pathogens. Collectively, this explains how the IMD pathway can be activated in the absence of canonical pathway initiators. Given that the UPR is highly conserved, UPR-initiated immunity may be a fundamental principle impacting vector competency across arthropods.
Collapse
Affiliation(s)
- Lindsay C. Sidak-Loftis
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Kristin L. Rosche
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Natasha Pence
- Institute of Biological Chemistry, Washington State University, Pullman, Washington, USA
| | - Jessica K. Ujczo
- United States Department of Agriculture, Agricultural Research Service, Animal Disease Research Unit, Pullman, Washington, USA
| | - Joanna Hurtado
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Elis A. Fisk
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Alan G. Goodman
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Susan M. Noh
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
- United States Department of Agriculture, Agricultural Research Service, Animal Disease Research Unit, Pullman, Washington, USA
| | - John W. Peters
- Institute of Biological Chemistry, Washington State University, Pullman, Washington, USA
| | - Dana K. Shaw
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| |
Collapse
|
28
|
Salcedo-Porras N, Oliveira PL, Guarneri AA, Lowenberger C. A fat body transcriptome analysis of the immune responses of Rhodnius prolixus to artificial infections with bacteria. Parasit Vectors 2022; 15:269. [PMID: 35906633 PMCID: PMC9335980 DOI: 10.1186/s13071-022-05358-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 06/10/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Rhodnius prolixus is an important vector of Trypanosoma cruzi, the causal agent of Chagas disease in humans. Despite the medical importance of this and other triatomine vectors, the study of their immune responses has been limited to a few molecular pathways and processes. Insect immunity studies were first described for holometabolous insects such as Drosophila melanogaster, and it was assumed that their immune responses were conserved in all insects. However, study of the immune responses of triatomines and other hemimetabolous insects has revealed discrepancies between these and the Drosophila model. METHODS To expand our understanding of innate immune responses of triatomines to pathogens, we injected fifth instar nymphs of R. prolixus with the Gram-negative (Gr-) bacterium Enterobacter cloacae, the Gram-positive (Gr+) bacterium Staphylococcus aureus, or phosphate-buffered saline (PBS), and evaluated transcript expression in the fat body 8 and 24 h post-injection (hpi). We analyzed the differential expression of transcripts at each time point, and across time, for each treatment. RESULTS At 8 hpi, the Gr- bacteria-injected group had a large number of differentially expressed (DE) transcripts, and most of the changes in transcript expression were maintained at 24 hpi. In the Gr+ bacteria treatment, few DE transcripts were detected at 8 hpi, but a large number of transcripts were DE at 24 hpi. Unexpectedly, the PBS control also had a large number of DE transcripts at 24 hpi. Very few DE transcripts were common to the different treatments and time points, indicating a high specificity of the immune responses of R. prolixus to different pathogens. Antimicrobial peptides known to be induced by the immune deficiency pathway were induced upon Gr- bacterial infection. Many transcripts of genes from the Toll pathway that are thought to participate in responses to Gr+ bacteria and fungi were induced by both bacteria and PBS treatment. Pathogen recognition receptors and serine protease cascade transcripts were also overexpressed after Gr- bacteria and PBS injections. Gr- injection also upregulated transcripts involved in the metabolism of tyrosine, a major substrate involved in the melanotic encapsulation response to pathogens. CONCLUSIONS These results reveal time-dependent pathogen-specific regulation of immune responses in triatomines, and hint at strong interactions between the immune deficiency and Toll pathways.
Collapse
Affiliation(s)
- Nicolas Salcedo-Porras
- Centre for Cell Biology, Development and Disease, Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6 Canada
| | - Pedro Lagerblad Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco D. Prédio do CCS, Ilha do Fundão, Rio de Janeiro, 21941-902 Brazil
| | - Alessandra Aparecida Guarneri
- Vector Behavior and Pathogen Interaction Group, Centro de Pesquisas René Rachou, Fiocruz, Avenida Augusto de Lima, 1715, Belo Horizonte, MG CEP 30190-009 Brazil
| | - Carl Lowenberger
- Centre for Cell Biology, Development and Disease, Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6 Canada
| |
Collapse
|
29
|
de la Fuente J, Contreras M. Additional considerations for anti-tick vaccine research. Expert Rev Vaccines 2022; 21:1019-1021. [PMID: 35475778 DOI: 10.1080/14760584.2022.2071704] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- José de la Fuente
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ciudad Real, Spain.,Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, USA
| | - Marinela Contreras
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ciudad Real, Spain
| |
Collapse
|
30
|
Wu-Chuang A, Hodžić A, Mateos-Hernández L, Estrada-Peña A, Obregon D, Cabezas-Cruz A. Current debates and advances in tick microbiome research. CURRENT RESEARCH IN PARASITOLOGY & VECTOR-BORNE DISEASES 2022; 1:100036. [PMID: 35284884 PMCID: PMC8906078 DOI: 10.1016/j.crpvbd.2021.100036] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/29/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022]
Abstract
The main importance of ticks resides in their ability to harbor pathogens that can be transmitted to terrestrial vertebrates including humans. Recently, studies have focused on the taxonomic and functional composition of the tick microbiome, its microbial diversity and variation under different factors including tick species, sex, and environment among others. Of special interest are the interactions between the tick, the microbiome and pathogens since tick microbiome can influence pathogen colonization within the tick vector, and potentially, transmission to the vertebrate host. In this review, we tackled a synthesis on the growing field of tick microbiomes. We focus on the current state of tick microbiome research, addressing controversial and hotly debated topics and advances in the precise manipulation of tick microbiome. Furthermore, we discuss the innovative anti-tick microbiota vaccines as a possible tool for microbiome modulation and thus, control of tick-borne diseases. Deciphering tick-microbiome pathogen interactions can spur new strategies to control tick-borne diseases via modulation of tick microbiome. Whether the diversity observed in tick microbiomes concerns the biology or the methodology remains an open question. Tick immunity must play a major role in selecting ‘who stays and who leaves’ the microbiome. Anti-tick microbiota vaccines can target specific bacteria and subsequently modulate tick microbiome.
Collapse
Affiliation(s)
- Alejandra Wu-Chuang
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, F-94700, France
| | - Adnan Hodžić
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210, Vienna, Austria
| | - Lourdes Mateos-Hernández
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, F-94700, France
| | | | - Dasiel Obregon
- School of Environmental Sciences University of Guelph, Guelph, Ontario, N1G 2W1, Canada
- Center for Nuclear Energy in Agriculture, University of São Paulo, Piracicaba, São Paulo, 13400-970, Brazil
| | - Alejandro Cabezas-Cruz
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, F-94700, France
- Corresponding author.
| |
Collapse
|
31
|
Hromníková D, Furka D, Furka S, Santana JAD, Ravingerová T, Klöcklerová V, Žitňan D. Prevention of tick-borne diseases: challenge to recent medicine. Biologia (Bratisl) 2022; 77:1533-1554. [PMID: 35283489 PMCID: PMC8905283 DOI: 10.1007/s11756-021-00966-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 11/10/2021] [Indexed: 12/14/2022]
Abstract
Abstract Ticks represent important vectors and reservoirs of pathogens, causing a number of diseases in humans and animals, and significant damage to livestock every year. Modern research into protection against ticks and tick-borne diseases focuses mainly on the feeding stage, i.e. the period when ticks take their blood meal from their hosts during which pathogens are transmitted. Physiological functions in ticks, such as food intake, saliva production, reproduction, development, and others are under control of neuropeptides and peptide hormones which may be involved in pathogen transmission that cause Lyme borreliosis or tick-borne encephalitis. According to current knowledge, ticks are not reservoirs or vectors for the spread of COVID-19 disease. The search for new vaccination methods to protect against ticks and their transmissible pathogens is a challenge for current science in view of global changes, including the increasing migration of the human population. Highlights • Tick-borne diseases have an increasing incidence due to climate change and increased human migration • To date, there is no evidence of transmission of coronavirus COVID-19 by tick as a vector • To date, there are only a few modern, effective, and actively- used vaccines against ticks or tick-borne diseases • Neuropeptides and their receptors expressed in ticks may be potentially used for vaccine design
Collapse
Affiliation(s)
- Dominika Hromníková
- Department of Molecular Physiology, Slovak Academy of Sciences, Institute of Zoology, Dúbravská cesta 9, 84506 Bratislava, Slovakia
| | - Daniel Furka
- Faculty of Natural Sciences, Department of Physical and Theoretical Chemistry, Comenius University, Mlynská dolina, Ilkovičova 6, 84104 Bratislava, SK Slovakia
- Department of Cardiovascular Physiology and Pathophysiology, Slovak Academy of Sciences, Institute of Heart Research, Dúbravská cesta 9, SK 84005 Bratislava, Slovakia
| | - Samuel Furka
- Faculty of Natural Sciences, Department of Physical and Theoretical Chemistry, Comenius University, Mlynská dolina, Ilkovičova 6, 84104 Bratislava, SK Slovakia
- Department of Cardiovascular Physiology and Pathophysiology, Slovak Academy of Sciences, Institute of Heart Research, Dúbravská cesta 9, SK 84005 Bratislava, Slovakia
| | - Julio Ariel Dueñas Santana
- Chemical Engineering Department, University of Matanzas, Km 3 Carretera a Varadero, 44740 Matanzas, CU Cuba
| | - Táňa Ravingerová
- Department of Cardiovascular Physiology and Pathophysiology, Slovak Academy of Sciences, Institute of Heart Research, Dúbravská cesta 9, SK 84005 Bratislava, Slovakia
| | - Vanda Klöcklerová
- Department of Molecular Physiology, Slovak Academy of Sciences, Institute of Zoology, Dúbravská cesta 9, 84506 Bratislava, Slovakia
| | - Dušan Žitňan
- Department of Molecular Physiology, Slovak Academy of Sciences, Institute of Zoology, Dúbravská cesta 9, 84506 Bratislava, Slovakia
| |
Collapse
|
32
|
Ring K, Couper LI, Sapiro AL, Yarza F, Yang XF, Clay K, Mateusiak C, Chou S, Swei A. Host blood meal identity modifies vector gene expression and competency. Mol Ecol 2022; 31:2698-2711. [PMID: 35231145 PMCID: PMC9314864 DOI: 10.1111/mec.16413] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 02/03/2022] [Accepted: 02/16/2022] [Indexed: 11/30/2022]
Abstract
A vector's susceptibility and ability to transmit a pathogen—termed vector competency—determines disease outcomes, yet the ecological factors influencing tick vector competency remain largely unknown. Ixodes pacificus, the tick vector of Borrelia burgdorferi (Bb) in the western U.S., feeds on rodents, birds, and lizards. Rodents and birds are reservoirs for Bb and infect juvenile ticks, while lizards are refractory to Bb and cannot infect feeding ticks. Additionally, the lizard bloodmeal contains borreliacidal properties, clearing previously infected feeding ticks of their Bb infection. Despite I. pacificus feeding on a range of hosts, it is undetermined how the host identity of the larval bloodmeal affects future nymphal vector competency. We experimentally evaluate the influence of larval host bloodmeal on Bb acquisition by nymphal I. pacificus. Larval I. pacificus were fed on either lizards or mice and after molting, nymphs were fed on Bb‐infected mice. We found that lizard‐fed larvae were significantly more likely to become infected with Bb during their next bloodmeal than mouse‐fed larvae. We also conducted the first RNA‐seq analysis on whole‐bodied I. pacificus and found significant upregulation of tick antioxidants and antimicrobial peptides in the lizard‐fed group. Our results indicate that the lizard bloodmeal significantly alters vector competency and gene regulation in ticks, highlighting the importance of host bloodmeal identity in vector‐borne disease transmission and upends prior notions about the role of lizards in Lyme disease community ecology.
Collapse
Affiliation(s)
- Kacie Ring
- Department of Ecology, Evolution, and Marine Biology, University of California, Santa Barbara, 93106
| | - Lisa I Couper
- Department of Biology, Stanford University, 327 Campus Drive, Stanford, 94305
| | - Anne L Sapiro
- Department of Biochemistry and Biophysics, University of California, San Francisco, 600 16th Street, 94158
| | - Fauna Yarza
- Department of Biochemistry and Biophysics, University of California, San Francisco, 600 16th Street, 94158
| | - X Frank Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, 635, Barnhill Drive, MS409J, 46202
| | - Keith Clay
- Department of Ecology and Evolutionary Biology, Tulane University, 6823 Charles Avenue, New Orleans, 70118
| | - Chase Mateusiak
- Center for Genome Science and Systems Biology, 4515 McKinley Ave, St. Louis, 63110
| | - Seemay Chou
- Department of Biochemistry and Biophysics, University of California, San Francisco, 600 16th Street, 94158.,Chan Zuckerberg Biohub, San Francisco, 94158
| | - Andrea Swei
- Department of Biology, San Francisco State University, 1600 Holloway Ave, San Francisco, 94132
| |
Collapse
|
33
|
Zhou K, Bai L, Nan X, Zhao K, Song Y, Li W, Wang Q. FADD regulates antibacterial immune responses via the immune deficiency signaling pathway in the Chinese mitten crab. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 128:104326. [PMID: 34856310 DOI: 10.1016/j.dci.2021.104326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 06/13/2023]
Abstract
In invertebrates, innate immune responses are the only defense against invading pathogens. The immune deficiency (IMD) signaling pathway protects invertebrates from bacterial infection by secreting antimicrobial peptides (AMPs). Fas-associated protein with death domain (FADD) activates AMPs and triggers apoptosis. However, FADD's function in crustaceans is unclear. Herein, the full-length FADD cDNA (EsFADD) was cloned from the Chinese mitten crab, Eriocheir sinensis. Vibrio parahaemolyticus infection upregulated EsFADD expression markedly. Knockdown of EsFADD in hemocytes suppressed the cytoplasm-to-nucleus translocation of transcription factor Relish under V. parahaemolyticus stimulation, which in turn reduced the expression of several AMPs. In vivo, silencing of EsFADD rendered crabs susceptible to bacterial infection and impaired their bacterial clearance. The results suggest that EsFADD is indispensable in IMD signal transduction in E. sinensis. In contrast to Drosophila, EsFADD barely promoted apoptosis. Our findings revealed the evolutionary conservation of FADD in crustaceans and provided insights into IMD signaling in invertebrates.
Collapse
Affiliation(s)
- Kaimin Zhou
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Longwei Bai
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xingyu Nan
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Ke Zhao
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yu Song
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Weiwei Li
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Qun Wang
- Laboratory of Invertebrate Immunological Defense & Reproductive Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
34
|
Wang XR, Cull B. Apoptosis and Autophagy: Current Understanding in Tick–Pathogen Interactions. Front Cell Infect Microbiol 2022; 12:784430. [PMID: 35155277 PMCID: PMC8829008 DOI: 10.3389/fcimb.2022.784430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/11/2022] [Indexed: 11/13/2022] Open
Abstract
Tick-borne diseases are a significant threat to human and animal health throughout the world. How tick-borne pathogens successfully infect and disseminate in both their vertebrate and invertebrate hosts is only partially understood. Pathogens have evolved several mechanisms to combat host defense systems, and to avoid and modulate host immunity during infection, therefore benefitting their survival and replication. In the host, pathogens trigger responses from innate and adaptive immune systems that recognize and eliminate invaders. Two important innate defenses against pathogens are the programmed cell death pathways of apoptosis and autophagy. This Mini Review surveys the current knowledge of apoptosis and autophagy pathways in tick-pathogen interactions, as well as the strategies evolved by pathogens for their benefit. We then assess the limitations to studying both pathways and discuss their participation in the network of the tick immune system, before highlighting future perspectives in this field. The knowledge gained would significantly enhance our understanding of the defense responses in vector ticks that regulate pathogen infection and burden, and form the foundation for future research to identify novel approaches to the control of tick-borne diseases.
Collapse
Affiliation(s)
- Xin-Ru Wang
- *Correspondence: Xin-Ru Wang, ; Benjamin Cull,
| | | |
Collapse
|
35
|
Viglietta M, Bellone R, Blisnick AA, Failloux AB. Vector Specificity of Arbovirus Transmission. Front Microbiol 2021; 12:773211. [PMID: 34956136 PMCID: PMC8696169 DOI: 10.3389/fmicb.2021.773211] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/19/2021] [Indexed: 12/20/2022] Open
Abstract
More than 25% of human infectious diseases are vector-borne diseases (VBDs). These diseases, caused by pathogens shared between animals and humans, are a growing threat to global health with more than 2.5 million annual deaths. Mosquitoes and ticks are the main vectors of arboviruses including flaviviruses, which greatly affect humans. However, all tick or mosquito species are not able to transmit all viruses, suggesting important molecular mechanisms regulating viral infection, dissemination, and transmission by vectors. Despite the large distribution of arthropods (mosquitoes and ticks) and arboviruses, only a few pairings of arthropods (family, genus, and population) and viruses (family, genus, and genotype) successfully transmit. Here, we review the factors that might limit pathogen transmission: internal (vector genetics, immune responses, microbiome including insect-specific viruses, and coinfections) and external, either biotic (adult and larvae nutrition) or abiotic (temperature, chemicals, and altitude). This review will demonstrate the dynamic nature and complexity of virus–vector interactions to help in designing appropriate practices in surveillance and prevention to reduce VBD threats.
Collapse
Affiliation(s)
- Marine Viglietta
- Unit of Arboviruses and Insect Vectors, Institut Pasteur, Sorbonne Université, Paris, France
| | - Rachel Bellone
- Unit of Arboviruses and Insect Vectors, Institut Pasteur, Sorbonne Université, Paris, France
| | - Adrien Albert Blisnick
- Unit of Arboviruses and Insect Vectors, Institut Pasteur, Sorbonne Université, Paris, France
| | - Anna-Bella Failloux
- Unit of Arboviruses and Insect Vectors, Institut Pasteur, Sorbonne Université, Paris, France
| |
Collapse
|
36
|
Aguilar-Díaz H, Quiroz-Castañeda RE, Salazar-Morales K, Cossío-Bayúgar R, Miranda-Miranda E. Tick Immunobiology and Extracellular Traps: An Integrative Vision to Control of Vectors. Pathogens 2021; 10:pathogens10111511. [PMID: 34832666 PMCID: PMC8621429 DOI: 10.3390/pathogens10111511] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/01/2021] [Accepted: 11/16/2021] [Indexed: 01/21/2023] Open
Abstract
Ticks are hematophagous ectoparasites that infest a diverse number of vertebrate hosts. The tick immunobiology plays a significant role in establishing and transmitting many pathogens to their hosts. To control tick infestations, the acaricide application is a commonly used method with severe environmental consequences and the selection of tick-resistant populations. With these drawbacks, new tick control methods need to be developed, and the immune system of ticks contains a plethora of potential candidates for vaccine design. Additionally, tick immunity is based on an orchestrated action of humoral and cellular immune responses. Therefore, the actors of these responses are the object of our study in this review since they are new targets in anti-tick vaccine design. We present their role in the immune response that positions them as feasible targets that can be blocked, inhibited, interfered with, and overexpressed, and then elucidate a new method to control tick infestations through the development of vaccines. We also propose Extracellular Traps Formation (ETosis) in ticks as a process to eliminate their natural enemies and those pathogens they transmit (vectorial capacity), which results attractive since they are a source of acting molecules with potential use as vaccines.
Collapse
Affiliation(s)
- Hugo Aguilar-Díaz
- Unidad de Artropodología, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad INIFAP, Jiutepec 62574, Mexico; (R.C.-B.); (E.M.-M.)
- Correspondence:
| | - Rosa Estela Quiroz-Castañeda
- Unidad de Anaplasmosis, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad INIFAP, Jiutepec 62574, Mexico;
| | - Karina Salazar-Morales
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico;
| | - Raquel Cossío-Bayúgar
- Unidad de Artropodología, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad INIFAP, Jiutepec 62574, Mexico; (R.C.-B.); (E.M.-M.)
| | - Estefan Miranda-Miranda
- Unidad de Artropodología, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad INIFAP, Jiutepec 62574, Mexico; (R.C.-B.); (E.M.-M.)
| |
Collapse
|
37
|
Salata C, Moutailler S, Attoui H, Zweygarth E, Decker L, Bell-Sakyi L. How relevant are in vitro culture models for study of tick-pathogen interactions? Pathog Glob Health 2021; 115:437-455. [PMID: 34190676 PMCID: PMC8635668 DOI: 10.1080/20477724.2021.1944539] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Although tick-borne infectious diseases threaten human and animal health worldwide, with constantly increasing incidence, little knowledge is available regarding vector-pathogen interactions and pathogen transmission. In vivo laboratory study of these subjects using live, intact ticks is expensive, labor-intensive, and challenging from the points of view of biosafety and ethics. Several in vitro models have been developed, including over 70 continuous cell lines derived from multiple tick species and a variety of tick organ culture systems, facilitating many research activities. However, some limitations have to be considered in the translation of the results from the in vitro environment to the in vivo situation of live, intact ticks, and vertebrate hosts. In this review, we describe the available in vitro models and selected results from their application to the study of tick-borne viruses, bacteria, and protozoa, where possible comparing these results to studies in live, intact ticks. Finally, we highlight the strengths and weaknesses of in vitro tick culture models and their essential role in tick-borne pathogen research.
Collapse
Affiliation(s)
- Cristiano Salata
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Sara Moutailler
- Laboratoire De Santé Animale, Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Maisons-Alfort, France
| | - Houssam Attoui
- Department of Animal Health, UMR1161 Virologie, INRAE, Ecole Nationale Vétérinaire d’Alfort, ANSES, Université Paris-Est, Maisons-Alfort, France
| | - Erich Zweygarth
- The Department of Veterinary Tropical Diseases, University of Pretoria, Pretoria, South Africa
| | - Lygia Decker
- Department of Preventive Veterinary Medicine, School of Veterinary Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Lesley Bell-Sakyi
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
38
|
Park JM, Oliva Chávez AS, Shaw DK. Ticks: More Than Just a Pathogen Delivery Service. Front Cell Infect Microbiol 2021; 11:739419. [PMID: 34540723 PMCID: PMC8440996 DOI: 10.3389/fcimb.2021.739419] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/11/2021] [Indexed: 12/17/2022] Open
Affiliation(s)
- Jason M Park
- Program in Vector-Borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, United States
| | - Adela S Oliva Chávez
- Department of Entomology, Texas A&M University, College Station, TX, United States
| | - Dana K Shaw
- Program in Vector-Borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, United States
| |
Collapse
|
39
|
Cell Line Platforms Support Research into Arthropod Immunity. INSECTS 2021; 12:insects12080738. [PMID: 34442304 PMCID: PMC8397109 DOI: 10.3390/insects12080738] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 12/21/2022]
Abstract
Simple Summary Many insect and tick species are serious pests, because insects damage crop plants and, along with ticks, transmit a wide range of human and animal diseases. One way of controlling these pests is by impairing their immune system, which protects them from bacterial, fungal, and viral infections. An important tool for studying immunity is using long-lasting cell cultures, known as cell lines. These lines can be frozen and thawed at will to be used in automated tests, and they provide consistent results over years. Questions that can be asked using cell lines include: How do insects or ticks recognize when they have been infected and by what organism? What kinds of defensive strategies do they use to contain or kill infectious agents? This article reviews research with insect or tick cell lines to answer these questions, as well as other questions relating to immunity. This review also discusses future research strategies for working with cell lines. Abstract Innate immune responses are essential to maintaining insect and tick health and are the primary defense against pathogenic viruses, bacteria, and fungi. Cell line research is a powerful method for understanding how invertebrates mount defenses against pathogenic organisms and testing hypotheses on how these responses occur. In particular, immortal arthropod cell lines are valuable tools, providing a tractable, high-throughput, cost-effective, and consistent platform to investigate the mechanisms underpinning insect and tick immune responses. The research results inform the controls of medically and agriculturally important insects and ticks. This review presents several examples of how cell lines have facilitated research into multiple aspects of the invertebrate immune response to pathogens and other foreign agents, as well as comments on possible future research directions in these robust systems.
Collapse
|
40
|
Cabrera-Jaramillo A, Monsalve S, Arroyave E, Rodas JD. Prevalence of Ehrlichia canis and Hepatozoon canis in sheltered dogs in southern Aburrá Valley, Colombia. REV COLOMB CIENC PEC 2021. [DOI: 10.17533/udea.rccp.v35n2a01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
41
|
Narasimhan S, Swei A, Abouneameh S, Pal U, Pedra JHF, Fikrig E. Grappling with the tick microbiome. Trends Parasitol 2021; 37:722-733. [PMID: 33962878 PMCID: PMC8282638 DOI: 10.1016/j.pt.2021.04.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/04/2021] [Accepted: 04/10/2021] [Indexed: 02/07/2023]
Abstract
Ixodes scapularis and Ixodes pacificus are the predominant vectors of multiple human pathogens, including Borrelia burgdorferi, one of the causative agents of Lyme disease in North America. Differences in the habitats and host preferences of these closely related tick species present an opportunity to examine key aspects of the tick microbiome. While advances in sequencing technologies have accelerated a descriptive understanding of the tick microbiome, molecular and mechanistic insights into the tick microbiome are only beginning to emerge. Progress is stymied by technical difficulties in manipulating the microbiome and by biological variables related to the life cycle of Ixodid ticks. This review highlights these challenges and examines avenues to understand the significance of the tick microbiome in tick biology.
Collapse
Affiliation(s)
- Sukanya Narasimhan
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06420, USA.
| | - Andrea Swei
- Department of Biology, San Francisco State University, San Francisco, CA 94132, USA
| | - Selma Abouneameh
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06420, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland School of Medicine, College Park, MD 20472, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 20472, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06420, USA
| |
Collapse
|
42
|
Function of cofactor Akirin2 in the regulation of gene expression in model human Caucasian neutrophil-like HL60 cells. Biosci Rep 2021; 41:229302. [PMID: 34291801 PMCID: PMC8298264 DOI: 10.1042/bsr20211120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 12/22/2022] Open
Abstract
The Akirin family of transcription cofactors are involved throughout the metazoan in the regulation of different biological processes (BPs) such as immunity, interdigital regression, muscle and neural development. Akirin do not have catalytic or DNA-binding capability and exert its regulatory function primarily through interacting proteins such as transcription factors, chromatin remodelers, and RNA-associated proteins. In the present study, we focused on the human Akirin2 regulome and interactome in neutrophil-like model human Caucasian promyelocytic leukemia HL60 cells. Our hypothesis is that metazoan evolved to have Akirin2 functional complements and different Akirin2-mediated mechanisms for the regulation of gene expression. To address this hypothesis, experiments were conducted using transcriptomics, proteomics and systems biology approaches in akirin2 knockdown and wildtype (WT) HL60 cells to characterize Akirin2 gene/protein targets, functional complements and to provide evidence of different mechanisms that may be involved in Akirin2-mediated regulation of gene expression. The results revealed Akirin2 gene/protein targets in multiple BPs with higher representation of immunity and identified immune response genes as candidate Akirin2 functional complements. In addition to linking chromatin remodelers with transcriptional activation, Akirin2 also interacts with histone H3.1 for regulation of gene expression.
Collapse
|
43
|
O'Neal AJ, Singh N, Mendes MT, Pedra JHF. The genus Anaplasma: drawing back the curtain on tick-pathogen interactions. Pathog Dis 2021; 79:ftab022. [PMID: 33792663 PMCID: PMC8062235 DOI: 10.1093/femspd/ftab022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022] Open
Abstract
Tick-borne illnesses pose a serious concern to human and veterinary health and their prevalence is on the rise. The interactions between ticks and the pathogens they carry are largely undefined. However, the genus Anaplasma, a group of tick-borne bacteria, has been instrumental in uncovering novel paradigms in tick biology. The emergence of sophisticated technologies and the convergence of entomology with microbiology, immunology, metabolism and systems biology has brought tick-Anaplasma interactions to the forefront of vector biology with broader implications for the infectious disease community. Here, we discuss the use of Anaplasma as an instrument for the elucidation of novel principles in arthropod-microbe interactions. We offer an outlook of the primary areas of study, outstanding questions and future research directions.
Collapse
Affiliation(s)
- Anya J O'Neal
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Nisha Singh
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Maria Tays Mendes
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
44
|
Artigas-Jerónimo S, Villar M, Cabezas-Cruz A, Caignard G, Vitour D, Richardson J, Lacour S, Attoui H, Bell-Sakyi L, Allain E, Nijhof AM, Militzer N, Pinecki Socias S, de la Fuente J. Tick Importin-α Is Implicated in the Interactome and Regulome of the Cofactor Subolesin. Pathogens 2021; 10:457. [PMID: 33920361 PMCID: PMC8069720 DOI: 10.3390/pathogens10040457] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/01/2021] [Accepted: 04/09/2021] [Indexed: 12/18/2022] Open
Abstract
Ticks and tick-borne diseases (TBDs) represent a burden for human and animal health worldwide. Currently, vaccines constitute the safest and most effective approach to control ticks and TBDs. Subolesin (SUB) has been identified as a vaccine antigen for the control of tick infestations and pathogen infection and transmission. The characterization of the molecular function of SUB and the identification of tick proteins interacting with SUB may provide the basis for the discovery of novel antigens and for the rational design of novel anti-tick vaccines. In the present study, we used the yeast two-hybrid system (Y2H) as an unbiased approach to identify tick SUB-interacting proteins in an Ixodes ricinus cDNA library, and studied the possible role of SUB as a chromatin remodeler through direct interaction with histones. The Y2H screening identified Importin-α as a potential SUB-interacting protein, which was confirmed in vitro in a protein pull-down assay. The sub gene expression levels in tick midgut and fat body were significantly higher in unfed than fed female ticks, however, the importin-α expression levels did not vary between unfed and fed ticks but tended to be higher in the ovary when compared to those in other organs. The effect of importin-α RNAi was characterized in I. ricinus under artificial feeding conditions. Both sub and importin-α gene knockdown was observed in all tick tissues and, while tick weight was significantly lower in sub RNAi-treated ticks than in controls, importin-α RNAi did not affect tick feeding or oviposition, suggesting that SUB is able to exert its function in the absence of Importin-α. Furthermore, SUB was shown to physically interact with histone 4, which was corroborated by protein pull-down and western blot analysis. These results confirm that by interacting with numerous tick proteins, SUB is a key cofactor of the tick interactome and regulome. Further studies are needed to elucidate the nature of the SUB-Importin-α interaction and the biological processes and functional implications that this interaction may have.
Collapse
Affiliation(s)
- Sara Artigas-Jerónimo
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (S.A.-J.); (M.V.)
| | - Margarita Villar
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (S.A.-J.); (M.V.)
- Biochemistry Section, Faculty of Science and Chemical Technologies, and Regional Centre for Biomedical Research (CRIB), University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - Alejandro Cabezas-Cruz
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, F-94700 Maisons-Alfort, France;
| | - Grégory Caignard
- UMR 1161 Virologie, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (G.C.); (D.V.); (J.R.); (S.L.); (H.A.); (E.A.)
| | - Damien Vitour
- UMR 1161 Virologie, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (G.C.); (D.V.); (J.R.); (S.L.); (H.A.); (E.A.)
| | - Jennifer Richardson
- UMR 1161 Virologie, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (G.C.); (D.V.); (J.R.); (S.L.); (H.A.); (E.A.)
| | - Sandrine Lacour
- UMR 1161 Virologie, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (G.C.); (D.V.); (J.R.); (S.L.); (H.A.); (E.A.)
| | - Houssam Attoui
- UMR 1161 Virologie, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (G.C.); (D.V.); (J.R.); (S.L.); (H.A.); (E.A.)
| | - Lesley Bell-Sakyi
- Tick Cell Biobank, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, 146 Brownlow Hill, Liverpool L3 5RF, UK;
| | - Eleonore Allain
- UMR 1161 Virologie, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (G.C.); (D.V.); (J.R.); (S.L.); (H.A.); (E.A.)
| | - Ard M. Nijhof
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany; (A.M.N.); (N.M.); (S.P.S.)
| | - Nina Militzer
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany; (A.M.N.); (N.M.); (S.P.S.)
| | - Sophia Pinecki Socias
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany; (A.M.N.); (N.M.); (S.P.S.)
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (S.A.-J.); (M.V.)
- Center for Veterinary Health Sciences, Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
45
|
Chen Y, Vargas SM, Smith TC, Karna SLR, MacMackin Ingle T, Wozniak KL, Wormley FL, Seshu J. Borrelia peptidoglycan interacting Protein (BpiP) contributes to the fitness of Borrelia burgdorferi against host-derived factors and influences virulence in mouse models of Lyme disease. PLoS Pathog 2021; 17:e1009535. [PMID: 33882111 PMCID: PMC8092773 DOI: 10.1371/journal.ppat.1009535] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 05/03/2021] [Accepted: 04/07/2021] [Indexed: 12/17/2022] Open
Abstract
The Peptidoglycan (PG) cell wall of the Lyme disease (LD) spirochete, Borrelia burgdorferi (Bb), contributes to structural and morphological integrity of Bb; is a persistent antigen in LD patients; and has a unique pentapeptide with L-Ornithine as the third amino acid that cross-links its glycan polymers. A borrelial homolog (BB_0167) interacted specifically with borrelilal PG via its peptidoglycan interacting motif (MHELSEKRARAIGNYL); was localized to the protoplasmic cylinder of Bb; and was designated as Borrelia peptidoglycan interacting Protein (BpiP). A bpiP mutant displayed no defect under in vitro growth conditions with similar levels of several virulence-related proteins. However, the burden of bpiP mutant in C3H/HeN mice at day 14, 28 and 62 post-infection was significantly lower compared to control strains. No viable bpiP mutant was re-isolated from any tissues at day 62 post-infection although bpiP mutant was able to colonize immunodeficient SCID at day 28 post-infection. Acquisition or transmission of bpiP mutant by Ixodes scapularis larvae or nymphs respectively, from and to mice, was significantly lower compared to control strains. Further analysis of bpiP mutant revealed increased sensitivity to vancomycin, osmotic stress, lysosomal extracts, human antimicrobial peptide cathelicidin-LL37, complement-dependent killing in the presence of day 14 post-infection mouse serum and increased internalization of CFSC-labeled bpiP mutant by macrophages and dendritic cells compared to control strains. These studies demonstrate the importance of accessory protein/s involved in sustaining integrity of PG and cell envelope during different phases of Bb infection.
Collapse
Affiliation(s)
- Yue Chen
- South Texas Center for Emerging Infectious Diseases (STCEID) and Department of Biology, The University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Sean M. Vargas
- South Texas Center for Emerging Infectious Diseases (STCEID) and Department of Biology, The University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Trever C. Smith
- South Texas Center for Emerging Infectious Diseases (STCEID) and Department of Biology, The University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Sai Lakshmi Rajasekhar Karna
- South Texas Center for Emerging Infectious Diseases (STCEID) and Department of Biology, The University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Taylor MacMackin Ingle
- South Texas Center for Emerging Infectious Diseases (STCEID) and Department of Biology, The University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Karen L. Wozniak
- South Texas Center for Emerging Infectious Diseases (STCEID) and Department of Biology, The University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Floyd L. Wormley
- South Texas Center for Emerging Infectious Diseases (STCEID) and Department of Biology, The University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Janakiram Seshu
- South Texas Center for Emerging Infectious Diseases (STCEID) and Department of Biology, The University of Texas at San Antonio, San Antonio, Texas, United States of America
| |
Collapse
|
46
|
Fogaça AC, Sousa G, Pavanelo DB, Esteves E, Martins LA, Urbanová V, Kopáček P, Daffre S. Tick Immune System: What Is Known, the Interconnections, the Gaps, and the Challenges. Front Immunol 2021; 12:628054. [PMID: 33737931 PMCID: PMC7962413 DOI: 10.3389/fimmu.2021.628054] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Ticks are ectoparasitic arthropods that necessarily feed on the blood of their vertebrate hosts. The success of blood acquisition depends on the pharmacological properties of tick saliva, which is injected into the host during tick feeding. Saliva is also used as a vehicle by several types of pathogens to be transmitted to the host, making ticks versatile vectors of several diseases for humans and other animals. When a tick feeds on an infected host, the pathogen reaches the gut of the tick and must migrate to its salivary glands via hemolymph to be successfully transmitted to a subsequent host during the next stage of feeding. In addition, some pathogens can colonize the ovaries of the tick and be transovarially transmitted to progeny. The tick immune system, as well as the immune system of other invertebrates, is more rudimentary than the immune system of vertebrates, presenting only innate immune responses. Although simpler, the large number of tick species evidences the efficiency of their immune system. The factors of their immune system act in each tick organ that interacts with pathogens; therefore, these factors are potential targets for the development of new strategies for the control of ticks and tick-borne diseases. The objective of this review is to present the prevailing knowledge on the tick immune system and to discuss the challenges of studying tick immunity, especially regarding the gaps and interconnections. To this end, we use a comparative approach of the tick immune system with the immune system of other invertebrates, focusing on various components of humoral and cellular immunity, such as signaling pathways, antimicrobial peptides, redox metabolism, complement-like molecules and regulated cell death. In addition, the role of tick microbiota in vector competence is also discussed.
Collapse
Affiliation(s)
- Andréa C. Fogaça
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Géssica Sousa
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniel B. Pavanelo
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Eliane Esteves
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Larissa A. Martins
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czechia
- Laboratory of Bacteriology, Tick-Pathogen Transmission Unit, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Veronika Urbanová
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Petr Kopáček
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Sirlei Daffre
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
47
|
Boulanger N, Wikel S. Induced Transient Immune Tolerance in Ticks and Vertebrate Host: A Keystone of Tick-Borne Diseases? Front Immunol 2021; 12:625993. [PMID: 33643313 PMCID: PMC7907174 DOI: 10.3389/fimmu.2021.625993] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/22/2021] [Indexed: 12/23/2022] Open
Abstract
Ticks and tick transmitted infectious agents are increasing global public health threats due to increasing abundance, expanding geographic ranges of vectors and pathogens, and emerging tick-borne infectious agents. Greater understanding of tick, host, and pathogen interactions will contribute to development of novel tick control and disease prevention strategies. Tick-borne pathogens adapt in multiple ways to very different tick and vertebrate host environments and defenses. Ticks effectively pharmacomodulate by its saliva host innate and adaptive immune defenses. In this review, we examine the idea that successful synergy between tick and tick-borne pathogen results in host immune tolerance that facilitates successful tick infection and feeding, creates a favorable site for pathogen introduction, modulates cutaneous and systemic immune defenses to establish infection, and contributes to successful long-term infection. Tick, host, and pathogen elements examined here include interaction of tick innate immunity and microbiome with tick-borne pathogens; tick modulation of host cutaneous defenses prior to pathogen transmission; how tick and pathogen target vertebrate host defenses that lead to different modes of interaction and host infection status (reservoir, incompetent, resistant, clinically ill); tick saliva bioactive molecules as important factors in determining those pathogens for which the tick is a competent vector; and, the need for translational studies to advance this field of study. Gaps in our understanding of these relationships are identified, that if successfully addressed, can advance the development of strategies to successfully disrupt both tick feeding and pathogen transmission.
Collapse
Affiliation(s)
- Nathalie Boulanger
- Fédération de Médecine Translationnelle - UR7290, Early Bacterial Virulence, Group Borrelia, Université de Strasbourg, Strasbourg, France.,Centre National de Référence Borrelia, Centre Hospitalier Universitaire, Strasbourg, France
| | - Stephen Wikel
- Department of Medical Sciences, Frank H. Netter, M.D., School of Medicine, Quinnipiac University, Hamden, CT, United States
| |
Collapse
|
48
|
Mateos-Hernández L, Pipová N, Allain E, Henry C, Rouxel C, Lagrée AC, Haddad N, Boulouis HJ, Valdés JJ, Alberdi P, de la Fuente J, Cabezas-Cruz A, Šimo L. Enlisting the Ixodes scapularis Embryonic ISE6 Cell Line to Investigate the Neuronal Basis of Tick-Pathogen Interactions. Pathogens 2021; 10:pathogens10010070. [PMID: 33466622 PMCID: PMC7828734 DOI: 10.3390/pathogens10010070] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 01/03/2023] Open
Abstract
Neuropeptides are small signaling molecules expressed in the tick central nervous system, i.e., the synganglion. The neuronal-like Ixodes scapularis embryonic cell line, ISE6, is an effective tool frequently used for examining tick–pathogen interactions. We detected 37 neuropeptide transcripts in the I. scapularis ISE6 cell line using in silico methods, and six of these neuropeptide genes were used for experimental validation. Among these six neuropeptide genes, the tachykinin-related peptide (TRP) of ISE6 cells varied in transcript expression depending on the infection strain of the tick-borne pathogen, Anaplasma phagocytophilum. The immunocytochemistry of TRP revealed cytoplasmic expression in a prominent ISE6 cell subpopulation. The presence of TRP was also confirmed in A. phagocytophilum-infected ISE6 cells. The in situ hybridization and immunohistochemistry of TRP of I. scapularis synganglion revealed expression in distinct neuronal cells. In addition, TRP immunoreaction was detected in axons exiting the synganglion via peripheral nerves as well as in hemal nerve-associated lateral segmental organs. The characterization of a complete Ixodes neuropeptidome in ISE6 cells may serve as an effective in vitro tool to study how tick-borne pathogens interact with synganglion components that are vital to tick physiology. Therefore, our current study is a potential stepping stone for in vivo experiments to further examine the neuronal basis of tick–pathogen interactions.
Collapse
Affiliation(s)
- Lourdes Mateos-Hernández
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (L.M.-H.); (E.A.); (C.R.); (A.-C.L.); (N.H.); (H.-J.B.)
| | - Natália Pipová
- Faculty of Science, Pavol Jozef Šafarik University in Košice, 04180 Košice, Slovakia;
| | - Eléonore Allain
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (L.M.-H.); (E.A.); (C.R.); (A.-C.L.); (N.H.); (H.-J.B.)
| | - Céline Henry
- AgroParisTech, Micalis Institute, Université Paris-Saclay, PAPPSO, INRAE, 78350 Jouy-en-Josas, France;
| | - Clotilde Rouxel
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (L.M.-H.); (E.A.); (C.R.); (A.-C.L.); (N.H.); (H.-J.B.)
| | - Anne-Claire Lagrée
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (L.M.-H.); (E.A.); (C.R.); (A.-C.L.); (N.H.); (H.-J.B.)
| | - Nadia Haddad
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (L.M.-H.); (E.A.); (C.R.); (A.-C.L.); (N.H.); (H.-J.B.)
| | - Henri-Jean Boulouis
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (L.M.-H.); (E.A.); (C.R.); (A.-C.L.); (N.H.); (H.-J.B.)
| | - James J. Valdés
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, 37005 Ceske Budejovice, Czech Republic;
- Department of Virology, Veterinary Research Institute, Hudcova 70, 62100 Brno, Czech Republic
| | - Pilar Alberdi
- SaBio Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (P.A.); (J.d.l.F.)
- Neuroplasticity and Neurodegeneration Group, Regional Centre for Biomedical Research (CRIB), Ciu-dad Real Medical School, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - José de la Fuente
- SaBio Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (P.A.); (J.d.l.F.)
- Center for Veterinary Health Sciences, Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078, USA
| | - Alejandro Cabezas-Cruz
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (L.M.-H.); (E.A.); (C.R.); (A.-C.L.); (N.H.); (H.-J.B.)
- Correspondence: (A.C.-C.); (L.Š.); Tel.: +33-6-31-23-51-91 (A.C.-C.); +33-1-49-77-46-52 (L.Š.)
| | - Ladislav Šimo
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (L.M.-H.); (E.A.); (C.R.); (A.-C.L.); (N.H.); (H.-J.B.)
- Correspondence: (A.C.-C.); (L.Š.); Tel.: +33-6-31-23-51-91 (A.C.-C.); +33-1-49-77-46-52 (L.Š.)
| |
Collapse
|
49
|
Helble JD, McCarthy JE, Hu LT. Interactions between Borrelia burgdorferi and its hosts across the enzootic cycle. Parasite Immunol 2021; 43:e12816. [PMID: 33368329 DOI: 10.1111/pim.12816] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/24/2022]
Abstract
The bacterial pathogen Borrelia burgdorferi is the causative agent of Lyme disease and is transmitted to humans through an Ixodes tick vector. B. burgdorferi is able to survive in both mammalian and tick hosts through careful modulation of its gene expression. This allows B. burgdorferi to adapt to the environmental and nutritional changes that occur when it is transmitted between the two hosts. Distinct interactions between the spirochete and its host occur at every step of the enzootic cycle and dictate the ability of the spirochete to survive until the next stage of the cycle. Studying the interface between B. burgdorferi, the Ixodes tick vector and the natural mammalian reservoirs has been made significantly more feasible through the complete genome sequences of the organisms and the advent of high throughput screening technologies. Ultimately, a thorough investigation of the interplay between the two domains (and two phyla within one domain) is necessary in order to completely understand how the pathogen is transmitted.
Collapse
Affiliation(s)
- Jennifer D Helble
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA, USA
| | - Julie E McCarthy
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA, USA
| | - Linden T Hu
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA, USA
| |
Collapse
|
50
|
Hart CE, Thangamani S. Tick-virus interactions: Current understanding and future perspectives. Parasite Immunol 2021; 43:e12815. [PMID: 33368375 DOI: 10.1111/pim.12815] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/30/2022]
Abstract
Ticks are the primary vector of arboviruses in temperate climates worldwide. They are both the vector of these pathogens to humans and an integral component of the viral sylvatic cycle. Understanding the tick-pathogen interaction provides information about the natural maintenance of these pathogens and informs the development of countermeasures against human infection. In this review, we discuss currently available information on tick-viral interactions within the broader scope of general tick immunology. While the tick immune response to several pathogens has been studied extensively, minimal work centres on responses to viral infection. This is largely due to the high pathogenicity of tick-borne viruses; this necessitates high-containment laboratories or low-pathogenicity substitute viruses. This has biased most research towards tick-borne flaviviruses. More work is required to fully understand the role of tick-virus interaction in sylvatic cycling and transmission of diverse tick-borne viruses.
Collapse
Affiliation(s)
- Charles Edward Hart
- Institute for Global Health and Translational Science, Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Saravanan Thangamani
- Institute for Global Health and Translational Science, Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|