1
|
Petit MJ, Flory C, Gu Q, Fares M, Lamont D, Score A, Davies K, Bell-Sakyi L, Scaturro P, Brennan B, Kohl A. Multi-omics analysis of SFTS virus infection in Rhipicephalus microplus cells reveals antiviral tick factors. Nat Commun 2025; 16:4732. [PMID: 40399277 PMCID: PMC12095547 DOI: 10.1038/s41467-025-59565-w] [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: 06/07/2024] [Accepted: 04/25/2025] [Indexed: 05/23/2025] Open
Abstract
The increasing prevalence of tick-borne arboviral infections worldwide necessitates advanced control strategies, particularly those targeting vectors, to mitigate the disease burden. However, the cellular interactions between arboviruses and ticks, especially for negative-strand RNA viruses, remain largely unexplored. Here, we employ a proteomics informed by transcriptomics approach to elucidate the cellular response of the Rhipicephalus microplus-derived BME/CTVM6 cell line to severe fever with thrombocytopenia syndrome virus (SFTSV) infection. We generate the de novo transcriptomes and proteomes of SFTSV- and mock-infected tick cells, identifying key host responses and regulatory pathways. Additionally, interactome analysis of the viral nucleoprotein (N) integrated host responses with viral replication and dsRNA-mediated gene silencing screen reveals two anti-SFTSV effectors: the N interacting RNA helicases DHX9 and UPF1. Collectively, our results provide insights into the antiviral responses of R. microplus vector cells and highlight critical SFTSV restriction factors, while enriching transcriptomic and proteomic resources for future research.
Collapse
Affiliation(s)
- Marine J Petit
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK.
- Microbes, Infection & Immunity, School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK.
| | | | - Quan Gu
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Mazigh Fares
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Douglas Lamont
- Fingerprints Proteomics Facility, School of Life Science, University of Dundee, Dundee, UK
| | - Alan Score
- Fingerprints Proteomics Facility, School of Life Science, University of Dundee, Dundee, UK
| | - Kelsey Davies
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Lesley Bell-Sakyi
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | | | - Benjamin Brennan
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK.
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK.
- Departments of Tropical Disease Biology and Vector Biology, Centre for Neglected Tropical Diseases, Liverpool School of Tropical Medicine, Liverpool, UK.
| |
Collapse
|
2
|
Cacace A, De Leva G, Di Lelio I, Becchimanzi A. Immune-Related Genes in the Honey Bee Mite Varroa destructor ( Acarina, Parasitidae). INSECTS 2025; 16:356. [PMID: 40332846 PMCID: PMC12027997 DOI: 10.3390/insects16040356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/24/2025] [Indexed: 05/08/2025]
Abstract
Despite its ecological and economic importance, many aspects of Varroa destructor's biology remain poorly understood, particularly its defense mechanisms against pathogens. The limited knowledge of Varroa's immunity has hindered the development of RNA interference (RNAi)-based strategies targeting immune-related genes. In this study, we investigated the immune gene repertoire of V. destructor by querying its NCBI nr protein database and comparing it to model species of ticks (Ixodes scapularis) and mites (Galendromus occidentalis and Tetranychus urticae). Transcription of candidate immune genes was confirmed by analyzing a de novo assembled transcriptome of V. destructor. Our findings reveal that V. destructor shares key immunological traits with ticks, including lysozymes, chitinases, and thioester-containing proteins (TEPs), but also shares the absence of transmembrane peptidoglycan recognition proteins (PGRPs), Gram-negative binding proteins, and several lectin families involved in pathogen recognition. Additionally, Varroa mites, like ticks, lack homologs of crucial immune signaling components, such as the unpaired ligand (JAK/STAT), Eiger (JNK), and multiple elements of the IMD pathway. They also do not encode canonical antimicrobial peptides (AMPs) like defensins but possess putative homologs of ctenidins, AMPs previously identified in spiders and ticks, which may be adopted as a novel genetic readout for immune response in mites. Our findings lay the groundwork for future functional studies on mite immunity and open new avenues for RNAi-based biocontrol strategies targeting immune pathways to enhance Varroa management.
Collapse
Affiliation(s)
- Alfonso Cacace
- Department of Agricultural Sciences, University of Naples Federico II, 80126 Naples, Italy; (A.C.); (G.D.L.); (I.D.L.)
| | - Giovanna De Leva
- Department of Agricultural Sciences, University of Naples Federico II, 80126 Naples, Italy; (A.C.); (G.D.L.); (I.D.L.)
| | - Ilaria Di Lelio
- Department of Agricultural Sciences, University of Naples Federico II, 80126 Naples, Italy; (A.C.); (G.D.L.); (I.D.L.)
- BAT Center—Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80126 Naples, Italy
| | - Andrea Becchimanzi
- Department of Agricultural Sciences, University of Naples Federico II, 80126 Naples, Italy; (A.C.); (G.D.L.); (I.D.L.)
- BAT Center—Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology, University of Naples Federico II, 80126 Naples, Italy
| |
Collapse
|
3
|
YUAN C, XU Q, NING Y, XIA Q. Potential mechanisms implied in tick infection by arboviruses and their transmission to vertebrate hosts. Integr Zool 2025; 20:315-330. [PMID: 39016029 PMCID: PMC11897945 DOI: 10.1111/1749-4877.12875] [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] [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 MedicineHainan Medical UniversityHaikouChina
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Center for Biosafety Mega‐ScienceChinese Academy of SciencesWuhanChina
| | - Qiong XU
- NHC Key Laboratory of Tropical Disease Control, School of Tropical MedicineHainan Medical UniversityHaikouChina
| | - Yunjia NING
- State Key Laboratory of Virology and National Virus Resource Center, Wuhan Institute of Virology, Center for Biosafety Mega‐ScienceChinese Academy of SciencesWuhanChina
- Hubei Jiangxia LaboratoryWuhanChina
| | - Qianfeng XIA
- NHC Key Laboratory of Tropical Disease Control, School of Tropical MedicineHainan Medical UniversityHaikouChina
| |
Collapse
|
4
|
Niu T, Yang B, Wang M, Wang Q, He D, Liu H, Li Y. Functional characterization and regulatory pattern of Neoseiulus barkeri peptidoglycan recognition protein (PGRP). Int J Biol Macromol 2025; 293:139458. [PMID: 39755301 DOI: 10.1016/j.ijbiomac.2025.139458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/27/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
Compared with traditional biological control, the co-use of entomopathogenic fungi and multiple enemies has made great progress in biocontrol technology. However, the risk posed by entomopathogenic fungi to their host has not been fully evaluated. Further, the interaction between them has not described adequately. In this study, we investigated the crucial role of peptidoglycan recognition protein (PGRP), an important pattern recognition receptor, involved in the resistance of Neoseiulus barkeri against the entomopathogenic fungus, Beauveria bassiana. Results showed that N. barkeri possessed one NbPGRP gene, which was mainly enriched in the ventral cuticle, and was significantly upregulated after B. bassiana induction. Knocking down NbPGRP resulted in decreased survival rate of mites and increased B. bassiana load. Also, recombinant NbPGRP inhibited the proliferation of B. bassiana, but also attached to spores by binding to surface PAMPs. Furthermore, the transcriptomics after NbPGRP silencing indicates that NbPGRP has a potential regulatory role in mite physiologies. These results showed that NbPGRP has a dual function of inhibiting the proliferation of entomopathogenic fungi and regulating mite resistance upon recognition of entomopathogens. This enhances understanding of the interaction mechanism between entomopathogens and their insect hosts.
Collapse
Affiliation(s)
- Tiandi Niu
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River, Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China; Yibin Academy of Southwest University, Yibin 644000, China
| | - Bowei Yang
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River, Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China; Yibin Academy of Southwest University, Yibin 644000, China
| | - Mian Wang
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River, Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China; Yibin Academy of Southwest University, Yibin 644000, China
| | - Qian Wang
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River, Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China; Yibin Academy of Southwest University, Yibin 644000, China
| | - Danyang He
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River, Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China; Yibin Academy of Southwest University, Yibin 644000, China
| | - Huai Liu
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River, Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China; Yibin Academy of Southwest University, Yibin 644000, China.
| | - Yaying Li
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River, Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China; Yibin Academy of Southwest University, Yibin 644000, China.
| |
Collapse
|
5
|
Vimonish R, Capelli-Peixoto J, Johnson W, Kappmeyer L, Saelao P, Taus N, Chung C, Ueti M. Transcriptomic analysis of Rhipicephalus microplus hemocytes from female ticks infected with Babesia bovis or Babesia bigemina. Parasit Vectors 2025; 18:37. [PMID: 39901199 PMCID: PMC11789329 DOI: 10.1186/s13071-025-06662-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/07/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Tick hemolymph is a sterile fluid that carries nutrients to maintain tick health. The hemolymph creates a hostile environment for invaders including the destruction of microorganisms by its circulating hemocytes. However, Babesia parasites escape and disseminate to other organs through the hemolymph to continue their transmission life cycle. Still, it is unknown how tick hemocytes respond to B. bovis or B. bigemina infection. In this study, we conducted a transcriptomic analysis of hemocytes from female Rhipicephalus microplus ticks infected with Babesia parasites to understand how gene expression changes during parasite infection. METHODS During Babesia acute infection, female R. microplus ticks were fed on bovines to acquire parasites. Engorged females were collected and incubated to develop Babesia kinetes in tick hemolymph. The hemolymph was examined to identify ticks that were highly infected with Babesia kinetes. Hemocyte cells were collected from replete female ticks infected with Babesia bovis or Babesia bigemina to perform high-throughput RNA-sequencing (RNA-Seq) analysis. RESULTS This study identified major changes in the gene profile of tick hemocytes during Babesia infection. The main groups of hemocyte genes that were altered during Babesia infection were associated with metabolism, immunity, and cytoskeletal rearrangement. Upregulated genes were mainly involved in defense mechanisms, while downregulated genes were related to cell proliferation and apoptosis. However, the expression of hemocyte genes varied among Babesia species' infections, and it reflected the changes that occurred in the tick's physiology, including growth, reproduction, and skeletal muscle development. CONCLUSIONS The differential gene expression of R. microplus hemocytes revealed that genes highly regulated upon Babesia infection were related to metabolism, tick immunity, cell growth, apoptosis, development, metabolism, and reproduction. Additional research is necessary to further define the genes that exhibited varying expression levels in hemocytes during the infection. The findings of this study will enhance our understanding on how Babesia parasites survive in the hostile environment of ticks and perpetuate their transmission cycle, ultimately contributing to the spread of bovine babesiosis.
Collapse
Affiliation(s)
- Rubikah Vimonish
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.
| | - Janaina Capelli-Peixoto
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Wendell Johnson
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | | | - Perot Saelao
- Veterinary Pest Genetic Research Unit, USDA-ARS, Kerrville, TX, USA
| | - Naomi Taus
- Animal Disease Research Unit, USDA-ARS, Pullman, WA, USA
| | - Chungwon Chung
- Animal Disease Research Unit, USDA-ARS, Pullman, WA, USA
| | - Massaro Ueti
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
- Animal Disease Research Unit, USDA-ARS, Pullman, WA, USA
| |
Collapse
|
6
|
Szczotko M, Antunes S, Domingos A, Dmitryjuk M. Investigation of genes expression of the JAK/STAT signalling pathway and AMPs in the presence of Borrelia spirochetes in Ixodes ricinus. Sci Rep 2025; 15:2869. [PMID: 39843584 PMCID: PMC11754740 DOI: 10.1038/s41598-025-87506-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 01/20/2025] [Indexed: 01/24/2025] Open
Abstract
Multicellular animals need to control the spread of invading pathogens. This is a particular challenge for blood-feeding vectors such as ticks, which ingest large amounts of blood potentially laden with harmful microorganisms. Ticks have a basic innate immune system and protect themselves from infection through innate immune responses involving pathways such as Janus kinase (JAK) or the signalling transducer activator of transcription (STAT). Direct antimicrobial defence occurs through the rapid synthesis of numerous antimicrobial agents including antimicrobial peptides (AMPs). The tick Ixodes ricinus is one of the main vectors of the Lyme disease pathogen, the spirochete Borrelia burgdorferi sensu lato. Data suggest that the JAK/STAT signalling pathway controls the expression of AMPs and regulates the infection of the pathogen in the tick body. The innate immune system during the off-host period keeps the level of spirochete infection in check. Spirochetes may influence the innate immune response in ticks. Therefore, the aim of this study was to analyse the expression of the genes related to the JAK/STAT pathway and selected AMPs in questing ticks in which B. burgorferi s.l. was detected. In the ticks infected with spirochetes, overexpression of genes related to the JAK/STAT signalling pathway was observed in the case of STAM and SOCS genes. AMPs genes such as def1, ric, lzs were overexpressed with different expression patterns. The results obtained suggest that AMPs may be involved in infection management in ticks.
Collapse
Affiliation(s)
- Magdalena Szczotko
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, Olsztyn, 10-719, Poland.
| | - Sandra Antunes
- Global Health and Tropical Medicine (GHTM), Associate Laboratory in Translation and Innovation Towards Global Health (LA-REAL), Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), Rua da Junqueira 100, Lisbon, 1349-008, Portugal
| | - Ana Domingos
- Global Health and Tropical Medicine (GHTM), Associate Laboratory in Translation and Innovation Towards Global Health (LA-REAL), Instituto de Higiene e Medicina Tropical (IHMT), Universidade NOVA de Lisboa (UNL), Rua da Junqueira 100, Lisbon, 1349-008, Portugal
| | - Małgorzata Dmitryjuk
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, Olsztyn, 10-719, Poland
| |
Collapse
|
7
|
Fongsaran C, Verhoeve VI, Jirakanwisal K, Harris EK, Macaluso KR. Identification and characterization of a Relish-type NF-κB, DvRelish, in Dermacentor variabilis in response to Rickettsia rickettsii infection. Front Cell Infect Microbiol 2024; 14:1494450. [PMID: 39735256 PMCID: PMC11682715 DOI: 10.3389/fcimb.2024.1494450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/14/2024] [Indexed: 12/31/2024] Open
Abstract
Ixodid ticks serve as hosts and transmission vectors for several obligate intracellular bacteria, including members of the spotted fever group (SFG) of Rickettsia. Although ticks generate an immune response to bacterial insults, many of the signaling molecules associated with the response and how they may contribute to vector competence for Rickettsia are undefined. In this study, we isolated a full-length dvrelish transcript from Dermacentor variabilis, which encoded a Relish-type NF-κB. The presence of a canonical Rel homology domain (RHD) consistent with NF-κB proteins suggested a role in tick immune response for DvRelish. The expression of DvRelish was confirmed in tick tissues and fluorescent microscopy of tick hemocytes indicated increased expression following infection with Rickettsia as compared to a non-tick-borne bacterial pathogen. To further determine the effect of dvRelish gene knockdown on rickettsial infection, we used RNA interference-mediated gene knockdown in D. variabilis and demonstrated that transcription of dvRelish was decreased after 24 h post-injection of siRNA. We then assessed the response of D. variabilis when exposed to Rickettsia rickettsii and determined that transcription of dvRelish was inversely associated with rickettsial loads at 48 h post-exposure. Further studies are required to broaden the understanding of differential immune responses in ticks to SFG Rickettsia infection and elucidate the role played by the arthropod immune system in vector competence.
Collapse
|
8
|
Petit MJ, Johnson N, Mansfield KL. Vectorial dynamics underpinning current and future tick-borne virus emergence in Europe. J Gen Virol 2024; 105. [PMID: 39526891 DOI: 10.1099/jgv.0.002041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Tick-borne diseases pose a growing threat to human and animal health in Europe, with tick-borne encephalitis virus (TBEV) and Crimean-Congo haemorrhagic fever virus (CCHFV), vectored by Ixodes ricinus and Hyalomma marginatum, respectively, emerging as primary public health concerns. The ability of ticks to transmit pathogens to multiple hosts and maintain infections across life stages makes them highly efficient vectors. However, many aspects of tick ecology and vectorial capacity remain understudied. This review examines key factors contributing to the vectorial competence of European ticks and their associated viruses. We first explore the influence of climate change on vector and disease ecology, using TBEV and CCHFV as case studies. We then analyse the role of the tick antiviral response in shaping vector competence. By integrating these elements, this review aims to enhance our understanding of tick-borne viral diseases and support the development of public health strategies, particularly through the One Health framework, to mitigate their impact in Europe.
Collapse
Affiliation(s)
- Marine J Petit
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Nicholas Johnson
- Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
- Animal and Plant Health Agency, Addlestone KT15 3NB, UK
| | | |
Collapse
|
9
|
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
|
10
|
Lyu B, Li J, Niemeyer B, Anderson DM, Beerntsen B, Song Q. Integrative analysis highlights molecular and immune responses of tick Amblyomma americanum to Escherichia coli challenge. Front Cell Infect Microbiol 2023; 13:1236785. [PMID: 37583446 PMCID: PMC10424933 DOI: 10.3389/fcimb.2023.1236785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 07/10/2023] [Indexed: 08/17/2023] Open
Abstract
Ticks are ectoparasites that can transmit various pathogens capable of causing life-threatening illnesses in people and animals, making them a severe public health threat. Understanding how ticks respond to bacterial infection is crucial for deciphering their immune defense mechanisms and identifying potential targets for controlling tick-borne diseases. In this study, an in-depth transcriptome analysis was used to investigate the molecular and immune responses of Amblyomma americanum to infection caused by the microinjection of Escherichia coli. With an abundance of differentially expressed genes discovered at different times, the analysis demonstrated significant changes in gene expression profiles in response to E. coli challenge. Notably, we found alterations in crucial immune markers, including the antimicrobial peptides defensin and microplusin, suggesting they may play an essential role in the innate immune response. Furthermore, KEGG analysis showed that following E. coli exposure, a number of key enzymes, including lysosomal alpha-glucosidase, fibroblast growth factor, legumain, apoptotic protease-activating factor, etc., were altered, impacting the activity of the lysosome, mitogen-activated protein kinase, antigen processing and presentation, bacterial invasion, apoptosis, and the Toll and immune deficiency pathways. In addition to the transcriptome analysis, we constructed protein interaction networks to elucidate the molecular interactions underlying the tick's response to E. coli challenge. Hub genes were identified, and their functional enrichment provided insights into the regulation of cytoskeleton rearrangement, apoptotic processes, and kinase activity that may occur in infected cells. Collectively, the findings shed light on the potential immune responses in A. americanum that control E. coli infection.
Collapse
Affiliation(s)
- Bo Lyu
- Division of Plant Science and Technology, University of Missouri, Columbia, MO, United States
| | - Jingjing Li
- Division of Plant Science and Technology, University of Missouri, Columbia, MO, United States
| | - Brigid Niemeyer
- Division of Plant Science and Technology, University of Missouri, Columbia, MO, United States
| | - Deborah M. Anderson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Brenda Beerntsen
- Division of Plant Science and Technology, University of Missouri, Columbia, MO, United States
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Qisheng Song
- Division of Plant Science and Technology, University of Missouri, Columbia, MO, United States
| |
Collapse
|
11
|
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: 15] [Impact Index Per Article: 7.5] [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
|
12
|
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
|
13
|
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: 4.5] [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
|
14
|
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: 17] [Impact Index Per Article: 8.5] [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
|
15
|
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
|
16
|
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: 11] [Impact Index Per Article: 3.7] [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
|
17
|
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
|
18
|
Guizzo MG, Tirloni L, Gonzalez SA, Farber MD, Braz G, Parizi LF, Dedavid E Silva LA, da Silva Vaz I, Oliveira PL. Coxiella Endosymbiont of Rhipicephalus microplus Modulates Tick Physiology With a Major Impact in Blood Feeding Capacity. Front Microbiol 2022; 13:868575. [PMID: 35591999 PMCID: PMC9111531 DOI: 10.3389/fmicb.2022.868575] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/14/2022] [Indexed: 11/30/2022] Open
Abstract
In the past decade, metagenomics studies exploring tick microbiota have revealed widespread interactions between bacteria and arthropods, including symbiotic interactions. Functional studies showed that obligate endosymbionts contribute to tick biology, affecting reproductive fitness and molting. Understanding the molecular basis of the interaction between ticks and their mutualist endosymbionts may help to develop control methods based on microbiome manipulation. Previously, we showed that Rhipicephalus microplus larvae with reduced levels of Coxiella endosymbiont of R. microplus (CERM) were arrested at the metanymph life stage (partially engorged nymph) and did not molt into adults. In this study, we performed a transcriptomic differential analysis of the R. microplus metanymph in the presence and absence of its mutualist endosymbiont. The lack of CERM resulted in an altered expression profile of transcripts from several functional categories. Gene products such as DA-P36, protease inhibitors, metalloproteases, and evasins, which are involved in blood feeding capacity, were underexpressed in CERM-free metanymphs. Disregulation in genes related to extracellular matrix remodeling was also observed in the absence of the symbiont. Taken together, the observed alterations in gene expression may explain the blockage of development at the metanymph stage and reveal a novel physiological aspect of the symbiont-tick-vertebrate host interaction.
Collapse
Affiliation(s)
- Melina Garcia Guizzo
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD, United States.,Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucas Tirloni
- Tick-Pathogen Transmission Unit, Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Sergio A Gonzalez
- Instituto de Agrobiotecnologia y Biologia Molecular (IABIMO), INTA-CONICET, Hurlingham, Argentina
| | - Marisa D Farber
- Instituto de Agrobiotecnologia y Biologia Molecular (IABIMO), INTA-CONICET, Hurlingham, Argentina
| | - Glória Braz
- Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luís Fernando Parizi
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Itabajara da Silva Vaz
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Pedro L Oliveira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| |
Collapse
|
19
|
Kumar D, Downs LP, Embers M, Flynt AS, Karim S. Identification of microRNAs in the Lyme Disease Vector Ixodes scapularis. Int J Mol Sci 2022; 23:5565. [PMID: 35628370 PMCID: PMC9141961 DOI: 10.3390/ijms23105565] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 02/01/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs involved in many biological processes, including the immune pathways that control bacterial, parasitic, and viral infections. Pathogens probably modify host miRNAs to facilitate successful infection, so they might be useful targets for vaccination strategies. There are few data on differentially expressed miRNAs in the black-legged tick Ixodes scapularis after infection with Borrelia burgdorferi, the causative agent of Lyme disease in the United States. Small RNA sequencing and qRT-PCR analysis were used to identify and validate differentially expressed I. scapularis salivary miRNAs. Small RNA-seq yielded 133,465,828 (≥18 nucleotides) and 163,852,135 (≥18 nucleotides) small RNA reads from Borrelia-infected and uninfected salivary glands for downstream analysis using the miRDeep2 algorithm. As such, 254 miRNAs were identified across all datasets, 25 of which were high confidence and 51 low confidence known miRNAs. Further, 23 miRNAs were differentially expressed in uninfected and infected salivary glands: 11 were upregulated and 12 were downregulated upon pathogen infection. Gene ontology and network analysis of target genes of differentially expressed miRNAs predicted roles in metabolic, cellular, development, cellular component biogenesis, and biological regulation processes. Several Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, including sphingolipid metabolism; valine, leucine and isoleucine degradation; lipid transport and metabolism; exosome biogenesis and secretion; and phosphate-containing compound metabolic processes, were predicted as targets of differentially expressed miRNAs. A qRT-PCR assay was utilized to validate the differential expression of miRNAs. This study provides new insights into the miRNAs expressed in I. scapularis salivary glands and paves the way for their functional manipulation to prevent or treat B. burgdorferi infection.
Collapse
Affiliation(s)
- Deepak Kumar
- Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA; (D.K.); (A.S.F.)
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA;
| | - Latoyia P. Downs
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA;
| | - Monica Embers
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA;
| | - Alex Sutton Flynt
- Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA; (D.K.); (A.S.F.)
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA;
| | - Shahid Karim
- Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA; (D.K.); (A.S.F.)
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA;
| |
Collapse
|
20
|
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: 53] [Impact Index Per Article: 17.7] [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
|
21
|
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: 0.5] [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
|
22
|
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: 2.0] [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
|
23
|
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: 60] [Impact Index Per Article: 15.0] [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
|
24
|
Martins LA, Palmisano G, Cortez M, Kawahara R, de Freitas Balanco JM, Fujita A, Alonso BI, Barros-Battesti DM, Braz GRC, Tirloni L, Esteves E, Daffre S, Fogaça AC. The intracellular bacterium Rickettsia rickettsii exerts an inhibitory effect on the apoptosis of tick cells. Parasit Vectors 2020; 13:603. [PMID: 33261663 PMCID: PMC7706286 DOI: 10.1186/s13071-020-04477-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Rickettsia rickettsii is a tick-borne obligate intracellular bacterium that causes Rocky Mountain spotted fever, a life-threatening illness. To obtain an insight into the vector-pathogen interactions, we assessed the effects of infection with R. rickettsii on the proteome cells of the tick embryonic cell line BME26. METHODS The proteome of BME26 cells was determined by label-free high-performance liquid chromatography coupled with tandem mass spectrometry analysis. Also evaluated were the effects of infection on the activity of caspase-3, assessed by the hydrolysis of a synthetic fluorogenic substrate in enzymatic assays, and on the exposition of phosphatidyserine, evaluated by live-cell fluorescence microscopy after labeling with annexin-V. Finally, the effects of activation or inhibition of caspase-3 activity on the growth of R. rickettsii in BME26 cells was determined. RESULTS Tick proteins of different functional classes were modulated in a time-dependent manner by R. rickettsii infection. Regarding proteins involved in apoptosis, certain negative regulators were downregulated at the initial phase of the infection (6 h) but upregulated in the middle of the exponential phase of the bacterial growth (48 h). Microorganisms are known to be able to inhibit apoptosis of the host cell to ensure their survival and proliferation. We therefore evaluated the effects of infection on classic features of apoptotic cells and observed DNA fragmentation exclusively in noninfected cells. Moreover, both caspase-3 activity and phosphatidylserine exposition were lower in infected than in noninfected cells. Importantly, while the activation of caspase-3 exerted a detrimental effect on rickettsial proliferation, its inhibition increased bacterial growth. CONCLUSIONS Taken together, these results show that R. rickettsii modulates the proteome and exerts an inhibitory effect on apoptosis in tick cellsthat seems to be important to ensure cell colonization.
Collapse
Affiliation(s)
- Larissa Almeida Martins
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.,Rocky Mountain Laboratories, National Institutes of Health, National Institute of Allergy and Infectious Diseases, Hamilton, USA
| | - Giuseppe Palmisano
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Mauro Cortez
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Rebeca Kawahara
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.,Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia
| | | | - André Fujita
- Department of Computational Science, Institute of Mathematics and Statistics, University of São Paulo, São Paulo, SP, Brazil
| | - Beatriz Iglesias Alonso
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | - Gloria Regina Cardoso Braz
- Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Lucas Tirloni
- Rocky Mountain Laboratories, National Institutes of Health, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA
| | - Eliane Esteves
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Sirlei Daffre
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Andréa Cristina Fogaça
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
25
|
Tirloni L, Braz G, Nunes RD, Gandara ACP, Vieira LR, Assumpcao TC, Sabadin GA, da Silva RM, Guizzo MG, Machado JA, Costa EP, Santos D, Gomes HF, Moraes J, dos Santos Mota MB, Mesquita RD, de Souza Leite M, Alvarenga PH, Lara FA, Seixas A, da Fonseca RN, Fogaça AC, Logullo C, Tanaka AS, Daffre S, Oliveira PL, da Silva Vaz I, Ribeiro JMC. A physiologic overview of the organ-specific transcriptome of the cattle tick Rhipicephalus microplus. Sci Rep 2020. [DOI: 10.1246/nikkashi.1979.101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AbstractTo further obtain insights into the Rhipicephalus microplus transcriptome, we used RNA-seq to carry out a study of expression in (i) embryos; (ii) ovaries from partially and fully engorged females; (iii) salivary glands from partially engorged females; (iv) fat body from partially and fully engorged females; and (v) digestive cells from partially, and (vi) fully engorged females. We obtained > 500 million Illumina reads which were assembled de novo, producing > 190,000 contigs, identifying 18,857 coding sequences (CDS). Reads from each library were mapped back into the assembled transcriptome giving a view of gene expression in different tissues. Transcriptomic expression and pathway analysis showed that several genes related in blood digestion and host-parasite interaction were overexpressed in digestive cells compared with other tissues. Furthermore, essential genes for the cell development and embryogenesis were overexpressed in ovaries. Taken altogether, these data offer novel insights into the physiology of production and role of saliva, blood digestion, energy metabolism, and development with submission of 10,932 novel tissue/cell specific CDS to the NCBI database for this important tick species.
Collapse
|
26
|
A physiologic overview of the organ-specific transcriptome of the cattle tick Rhipicephalus microplus. Sci Rep 2020; 10:18296. [PMID: 33106528 PMCID: PMC7588415 DOI: 10.1038/s41598-020-75341-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/06/2020] [Indexed: 12/13/2022] Open
Abstract
To further obtain insights into the Rhipicephalus microplus transcriptome, we used RNA-seq to carry out a study of expression in (i) embryos; (ii) ovaries from partially and fully engorged females; (iii) salivary glands from partially engorged females; (iv) fat body from partially and fully engorged females; and (v) digestive cells from partially, and (vi) fully engorged females. We obtained > 500 million Illumina reads which were assembled de novo, producing > 190,000 contigs, identifying 18,857 coding sequences (CDS). Reads from each library were mapped back into the assembled transcriptome giving a view of gene expression in different tissues. Transcriptomic expression and pathway analysis showed that several genes related in blood digestion and host-parasite interaction were overexpressed in digestive cells compared with other tissues. Furthermore, essential genes for the cell development and embryogenesis were overexpressed in ovaries. Taken altogether, these data offer novel insights into the physiology of production and role of saliva, blood digestion, energy metabolism, and development with submission of 10,932 novel tissue/cell specific CDS to the NCBI database for this important tick species.
Collapse
|
27
|
Yuan C, Wu J, Peng Y, Li Y, Shen S, Deng F, Hu Z, Zhou J, Wang M, Zou Z. Transcriptome analysis of the innate immune system of Hyalomma asiaticum. J Invertebr Pathol 2020; 177:107481. [PMID: 33035534 DOI: 10.1016/j.jip.2020.107481] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/27/2020] [Accepted: 10/02/2020] [Indexed: 01/16/2023]
Abstract
Ticks are considered to be the second most important vectors of human infectious diseases. The innate immune system is the key factor that affects its vector competence. Hyalomma asiaticum is the primary vector of Crimean-Congo hemorrhagic fever virus (CCHFV). However, the immune system of H. asiaticum remains virtually unknown. Here, a high throughput full-length mRNA sequencing method was adopted to define the immunotranscriptome of H. asiaticum infected with the fungal pathogen Beauveria bassiana and gram-negative bacterium Enterobacter cloacae. The analysis yielded 22,300 isoforms with an average length of 3233 bps. In total, 68 potential immunity-related genes were identified based on similarity to the homologs known to be involved in immunity. These included most members of the Toll and JAK/STAT signaling pathways, but not the IMD signaling pathway. Moreover, two copies of Dicer-2 and five copies of Argonaute-2 were detected. These genes are postulated to be involved in the RNA interference (RNAi) pathway, which is an important defense against RNA viruses. Overall, this study provides the foundation for understanding the immune response of H. asiaticum to CCHFV.
Collapse
Affiliation(s)
- Chuanfei Yuan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China; State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jia Wu
- Wuhan National Biosafety Laboratory, Mega-Science Center for Bio-Safety Research, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yun Peng
- Wuhan National Biosafety Laboratory, Mega-Science Center for Bio-Safety Research, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yufeng Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Shu Shen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Fei Deng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Zhihong Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Jinlin Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China.
| | - Manli Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine and Laboratory Medicine, Hainan Medical University, Haikou 571199, China.
| |
Collapse
|
28
|
Abstract
Borrelia burgdorferi is the causative agent of Lyme disease and is transmitted to vertebrate hosts by Ixodes spp. ticks. The spirochaete relies heavily on its arthropod host for basic metabolic functions and has developed complex interactions with ticks to successfully colonize, persist and, at the optimal time, exit the tick. For example, proteins shield spirochaetes from immune factors in the bloodmeal and facilitate the transition between vertebrate and arthropod environments. On infection, B. burgdorferi induces selected tick proteins that modulate the vector gut microbiota towards an environment that favours colonization by the spirochaete. Additionally, the recent sequencing of the Ixodes scapularis genome and characterization of tick immune defence pathways, such as the JAK–STAT, immune deficiency and cross-species interferon-γ pathways, have advanced our understanding of factors that are important for B. burgdorferi persistence in the tick. In this Review, we summarize interactions between B. burgdorferi and I. scapularis during infection, as well as interactions with tick gut and salivary gland proteins important for establishing infection and transmission to the vertebrate host. Borrelia burgdorferi has a complex life cycle with several different hosts, causing Lyme disease when it infects humans. In this Review, Fikrig and colleagues discuss how B. burgdorferi infects and interacts with its tick vector to ensure onward transmission.
Collapse
|
29
|
Taking Insect Immunity to the Single-Cell Level. Trends Immunol 2020; 41:190-199. [DOI: 10.1016/j.it.2020.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 01/11/2020] [Accepted: 01/12/2020] [Indexed: 12/16/2022]
|
30
|
Salcedo-Porras N, Lowenberger C. The innate immune system of kissing bugs, vectors of chagas disease. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 98:119-128. [PMID: 31014953 DOI: 10.1016/j.dci.2019.04.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/09/2019] [Accepted: 04/18/2019] [Indexed: 05/08/2023]
Abstract
Kissing bugs have long served as models to study many aspects of insect physiology. They also serve as vectors for the parasite Trypanosoma cruzi that causes Chagas disease in humans. The overall success of insects is due, in part, to their ability to recognize parasites and pathogens as non-self and to eliminate them using their innate immune system. This immune system comprises physical barriers, cellular responses (phagocytosis, nodulation and encapsulation), and humoral factors (antimicrobial peptides and the prophenoloxidase cascade). Trypanosoma cruzi survives solely in the gastrointestinal (GI) tract of the vector; if it migrates to the hemocoel it is eliminated. Kissing bugs may not mount a vigorous immune response in the GI tract to avoid eliminating obligate symbiotic microbes on which they rely for survival. Here we describe the current knowledge of innate immunity in kissing bugs and new opportunities using genomic and transcriptomic approaches to study the complex triatomine-trypanosome-microbiome interactions.
Collapse
Affiliation(s)
- Nicolás Salcedo-Porras
- Department of Biological Sciences, Simon Fraser University, 8888 University Dr., Burnaby, V5A 1S6, BC, Canada.
| | - Carl Lowenberger
- Department of Biological Sciences, Simon Fraser University, 8888 University Dr., Burnaby, V5A 1S6, BC, Canada.
| |
Collapse
|
31
|
Sackton TB. Comparative genomics and transcriptomics of host-pathogen interactions in insects: evolutionary insights and future directions. CURRENT OPINION IN INSECT SCIENCE 2019; 31:106-113. [PMID: 31109663 DOI: 10.1016/j.cois.2018.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 12/21/2018] [Accepted: 12/22/2018] [Indexed: 06/09/2023]
Abstract
Classical evolutionary studies of protein-coding genes have established that genes in the canonical immune system are often among the most rapidly evolving within and between species. As more genomes and transcriptomes across insects are sequenced, it is becoming clear that duplications and losses of immune genes are also a likely consequence of host-pathogen interactions. Furthermore, particular species respond to diverse pathogenic challenges with a wide range of challenge-specific responses that are still poorly understood. Transcriptional studies, using RNA-seq to characterize the infection-regulated transcriptome of diverse insects, are crucial for additional progress in understanding the ecology and evolution of the full complexity of the host response.
Collapse
Affiliation(s)
- Timothy B Sackton
- Informatics Group, Faculty of Arts and Sciences, Harvard University, Cambridge, MA 02138, United States.
| |
Collapse
|
32
|
p47 licenses activation of the immune deficiency pathway in the tick Ixodes scapularis. Proc Natl Acad Sci U S A 2018; 116:205-210. [PMID: 30559180 DOI: 10.1073/pnas.1808905116] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The E3 ubiquitin ligase X-linked inhibitor of apoptosis (XIAP) acts as a molecular rheostat for the immune deficiency (IMD) pathway of the tick Ixodes scapularis How XIAP activates the IMD pathway in response to microbial infection remains ill defined. Here, we identified the XIAP enzymatic substrate p47 as a positive regulator of the I. scapularis IMD network. XIAP polyubiquitylates p47 in a lysine 63-dependent manner and interacts with the p47 ubiquitin-like (UBX) module. p47 also binds to Kenny (IKKγ/NEMO), the regulatory subunit of the inhibitor of nuclear factor (NF)- κB kinase complex. Replacement of the amino acid lysine to arginine within the p47 linker region completely abrogated molecular interactions with Kenny. Furthermore, mitigation of p47 transcription levels through RNA interference in I. scapularis limited Kenny accumulation, reduced phosphorylation of IKKβ (IRD5), and impaired cleavage of the NF-κB molecule Relish. Accordingly, disruption of p47 expression increased microbial colonization by the Lyme disease spirochete Borrelia burgdorferi and the rickettsial agent Anaplasma phagocytophilum Collectively, we highlight the importance of ticks for the elucidation of paradigms in arthropod immunology. Manipulating immune signaling cascades within I. scapularis may lead to innovative approaches to reducing the burden of tick-borne diseases.
Collapse
|
33
|
Vieira CS, Moreira OC, Batista KKS, Ratcliffe NA, Castro DP, Azambuja P. The NF-κB Inhibitor, IMD-0354, Affects Immune Gene Expression, Bacterial Microbiota and Trypanosoma cruzi Infection in Rhodnius prolixus Midgut. Front Physiol 2018; 9:1189. [PMID: 30233391 PMCID: PMC6128222 DOI: 10.3389/fphys.2018.01189] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 08/07/2018] [Indexed: 12/12/2022] Open
Abstract
Rhodnius prolixus is an insect vector of Trypanosoma cruzi, the causative agent of Chagas disease in Latin America. Nuclear factor-κB (NF-κB) transcription factors (TF) are conserved components of the innate immune system in several multicellular organisms including insects. The drug IMD-0354 [N-(3,5-bis-trifluoromethyl-phenyl)-5-chloro-2-hydroxy-benzamide] is a selective inhibitor of IκB kinases. It blocks IκBα phosphorylation thus preventing nuclear translocation of the NF-κb TF. In humans, NF-κB is involved in several biological processes such as inflammation, cell proliferation and immunity. In insects, the activation of the immune system upon microbial challenge can be controlled by signaling pathways such as the immune deficiency (IMD) and Toll, to combat infection. These activated pathways signal to downstream NF-κB TF to stimulate specific immune genes, triggering the synthesis of several molecules such as the antimicrobial peptides. In Drosophila melanogaster, the activation and regulation of NF-κB TF have been elucidated, while in triatomines these mechanisms are not fully understood Therefore, the present study investigated the effects of oral administration of the drug IMD-0354 on the R. prolixus immune response to challenge with bacteria and T. cruzi, as well as the impact on the gut bacterial microbiota. R. prolixus were fed with rabbit blood containing IMD-0354 and Escherichia coli, Staphylococcus aureus, or T. cruzi. The effects of IMD-0354 on insect mortality and antimicrobial activity in insect midgut samples, as well as the relative expression of R. prolixus immune genes were recorded. The bacterial microbiota was analyzed, and viable parasites were counted in insect midgut samples. The IMD-0354 treatment modulated antibacterial activity and the gene expression patterns of defensin A, defensin B, defensin C, and prolixicin, and the genes involved in the IMD and Toll pathways. Additionally, there was an increase of bacterial microbiota in treated insects. Insects treated with IMD-0354 and concomitantly infected with bacteria or T. cruzi through the blood meal had increased mortality, while the T. cruzi population in R. prolixus midgut was reduced. The inhibitory effect of IMD-0354 indicates the importance of NF-κB TF in the innate immune responses involved in the control of bacteria and parasite infections in the R. prolixus midgut.
Collapse
Affiliation(s)
- Cecilia S Vieira
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, Brazil
| | - Otacílio C Moreira
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, Brazil
| | - Kate K S Batista
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, Brazil
| | - Norman A Ratcliffe
- Laboratório de Biologia de Insetos, Universidade Federal Fluminense, Niterói, Brazil.,College of Science, Swansea University, Wales, United Kingdom
| | - Daniele P Castro
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, Brazil.,Departamento de Entomologia Molecular, Instituto Nacional de Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Patrícia Azambuja
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, Brazil.,Departamento de Entomologia Molecular, Instituto Nacional de Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| |
Collapse
|
34
|
Wang J, Song X, Wang M. Peptidoglycan recognition proteins in hematophagous arthropods. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 83:89-95. [PMID: 29269264 PMCID: PMC5889321 DOI: 10.1016/j.dci.2017.12.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/11/2017] [Accepted: 12/18/2017] [Indexed: 05/24/2023]
Abstract
Hematophagous arthropods are medically important disease vectors that transmit a variety of pathogens. Unlike mammals that employ both innate and adaptive immunity to clear invading pathogens, these vectors rely mainly on an innate immune system to combat pathogens. Peptidoglycan recognition proteins (PGRPs) are important components of innate immune signaling pathways and are responsible for recognizing microbe-associated molecular patterns (MAMPs), thus regulating host immune interactions with both harmful and helpful microbes. Here we review a number of recent studies in different vectors that address the function of PGRPs in immune regulation. Further, we discuss the variation of PGRPs between vectors and Drosophila.
Collapse
Affiliation(s)
- Jingwen Wang
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, PR China.
| | - Xiumei Song
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, PR China
| | - Mengfei Wang
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, PR China
| |
Collapse
|
35
|
Bell-Sakyi L, Darby A, Baylis M, Makepeace BL. The Tick Cell Biobank: A global resource for in vitro research on ticks, other arthropods and the pathogens they transmit. Ticks Tick Borne Dis 2018; 9:1364-1371. [PMID: 29886187 PMCID: PMC6052676 DOI: 10.1016/j.ttbdis.2018.05.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/18/2018] [Accepted: 05/30/2018] [Indexed: 12/11/2022]
Abstract
Tick cell lines are increasingly used in many fields of tick and tick-borne disease research. The Tick Cell Biobank was established in 2009 to facilitate the development and uptake of these unique and valuable resources. As well as serving as a repository for existing and new ixodid and argasid tick cell lines, the Tick Cell Biobank supplies cell lines and training in their maintenance to scientists worldwide and generates novel cultures from tick species not already represented in the collection. Now part of the Institute of Infection and Global Health at the University of Liverpool, the Tick Cell Biobank has embarked on a new phase of activity particularly targeted at research on problems caused by ticks, other arthropods and the diseases they transmit in less-developed, lower- and middle-income countries. We are carrying out genotypic and phenotypic characterisation of selected cell lines derived from tropical tick species. We continue to expand the culture collection, currently comprising 63 cell lines derived from 18 ixodid and argasid tick species and one each from the sand fly Lutzomyia longipalpis and the biting midge Culicoides sonorensis, and are actively engaging with collaborators to obtain starting material for primary cell cultures from other midge species, mites, tsetse flies and bees. Outposts of the Tick Cell Biobank will be set up in Malaysia, Kenya and Brazil to facilitate uptake and exploitation of cell lines and associated training by scientists in these and neighbouring countries. Thus the Tick Cell Biobank will continue to underpin many areas of global research into biology and control of ticks, other arthropods and vector-borne viral, bacterial and protozoan pathogens.
Collapse
Affiliation(s)
- Lesley Bell-Sakyi
- Institute of Infection and Global Health, University of Liverpool, Liverpool Science Park IC2, 146 Brownlow Hill, Liverpool L3 5RF, United Kingdom.
| | - Alistair Darby
- Institute of Integrative Biology, University of Liverpool, Biosciences Building, Crown Street, Liverpool L69 7ZB, United Kingdom.
| | - Matthew Baylis
- Institute of Infection and Global Health, University of Liverpool, Liverpool Science Park IC2, 146 Brownlow Hill, Liverpool L3 5RF, United Kingdom; NIHR Health Protection Research Institute in Emerging and Zoonotic Infections, Institute of Infection and Global Health, University of Liverpool, The Ronald Ross Building, 8 West Derby Street, Liverpool L69 7BE, United Kingdom.
| | - Benjamin L Makepeace
- Institute of Infection and Global Health, University of Liverpool, Liverpool Science Park IC2, 146 Brownlow Hill, Liverpool L3 5RF, United Kingdom.
| |
Collapse
|
36
|
Kitsou C, Pal U. Ixodes Immune Responses Against Lyme Disease Pathogens. Front Cell Infect Microbiol 2018; 8:176. [PMID: 29896452 PMCID: PMC5986905 DOI: 10.3389/fcimb.2018.00176] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/08/2018] [Indexed: 12/15/2022] Open
Abstract
Although Ixodes scapularis and other related tick species are considered prolific vectors for a number of important human diseases, many aspects of their biology, microbial interactions, and immunity are largely unknown; in particular, how these ancient vectors recognize invading pathogens like Borrelia burgdorferi and influence their persistence. The analysis of the Ixodes genome and a limited set of transcriptomic data have established that ticks encode many components of classical immune pathways; yet at the same time, they lack many key orthologs of these recognition networks. Therefore, whether a given immune pathway is active in Ixodes ticks and how precisely they exert its microbicidal functions are only incompletely delineated. A few recent studies have suggested that classical pathways like the Janus Kinase/Signal Transducer and Activator of Transcription (JAK/STAT) as well as immunodeficiency (IMD) pathways are fully functional in I. scapularis, and upon challenge with microbes, generate potent microbicidal responses against diverse tick-borne pathogens including B. burgdorferi. These studies also highlight novel concepts of vector immunity that include both a direct and an indirect mode of recognition of pathogens, as well as the influence of the gut microbiome, which ultimately dictates the outcome of a robust microbicidal response. Further understanding of how Ixodes ticks recognize and suppress invading microbes like B. burgdorferi will enrich our fundamental knowledge of vector immunobiology, thereby contributing to the development of future interventions to better control the tick-borne pathogen.
Collapse
Affiliation(s)
- Chrysoula Kitsou
- Department of Veterinary Medicine and Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD, United States
| | - Utpal Pal
- Department of Veterinary Medicine and Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD, United States
| |
Collapse
|
37
|
Vieira CS, Moreira OC, Batista KKS, Ratcliffe NA, Castro DP, Azambuja P. The NF-κB Inhibitor, IMD-0354, Affects Immune Gene Expression, Bacterial Microbiota and Trypanosoma cruzi Infection in Rhodnius prolixus Midgut. Front Physiol 2018. [PMID: 30233391 DOI: 10.3389/fphys.2018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2023] Open
Abstract
Rhodnius prolixus is an insect vector of Trypanosoma cruzi, the causative agent of Chagas disease in Latin America. Nuclear factor-κB (NF-κB) transcription factors (TF) are conserved components of the innate immune system in several multicellular organisms including insects. The drug IMD-0354 [N-(3,5-bis-trifluoromethyl-phenyl)-5-chloro-2-hydroxy-benzamide] is a selective inhibitor of IκB kinases. It blocks IκBα phosphorylation thus preventing nuclear translocation of the NF-κb TF. In humans, NF-κB is involved in several biological processes such as inflammation, cell proliferation and immunity. In insects, the activation of the immune system upon microbial challenge can be controlled by signaling pathways such as the immune deficiency (IMD) and Toll, to combat infection. These activated pathways signal to downstream NF-κB TF to stimulate specific immune genes, triggering the synthesis of several molecules such as the antimicrobial peptides. In Drosophila melanogaster, the activation and regulation of NF-κB TF have been elucidated, while in triatomines these mechanisms are not fully understood Therefore, the present study investigated the effects of oral administration of the drug IMD-0354 on the R. prolixus immune response to challenge with bacteria and T. cruzi, as well as the impact on the gut bacterial microbiota. R. prolixus were fed with rabbit blood containing IMD-0354 and Escherichia coli, Staphylococcus aureus, or T. cruzi. The effects of IMD-0354 on insect mortality and antimicrobial activity in insect midgut samples, as well as the relative expression of R. prolixus immune genes were recorded. The bacterial microbiota was analyzed, and viable parasites were counted in insect midgut samples. The IMD-0354 treatment modulated antibacterial activity and the gene expression patterns of defensin A, defensin B, defensin C, and prolixicin, and the genes involved in the IMD and Toll pathways. Additionally, there was an increase of bacterial microbiota in treated insects. Insects treated with IMD-0354 and concomitantly infected with bacteria or T. cruzi through the blood meal had increased mortality, while the T. cruzi population in R. prolixus midgut was reduced. The inhibitory effect of IMD-0354 indicates the importance of NF-κB TF in the innate immune responses involved in the control of bacteria and parasite infections in the R. prolixus midgut.
Collapse
Affiliation(s)
- Cecilia S Vieira
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, Brazil
| | - Otacílio C Moreira
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, Brazil
| | - Kate K S Batista
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, Brazil
| | - Norman A Ratcliffe
- Laboratório de Biologia de Insetos, Universidade Federal Fluminense, Niterói, Brazil
- College of Science, Swansea University, Wales, United Kingdom
| | - Daniele P Castro
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, Brazil
- Departamento de Entomologia Molecular, Instituto Nacional de Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Patrícia Azambuja
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, Brazil
- Departamento de Entomologia Molecular, Instituto Nacional de Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| |
Collapse
|
38
|
Immune-related redox metabolism of embryonic cells of the tick Rhipicephalus microplus (BME26) in response to infection with Anaplasma marginale. Parasit Vectors 2017; 10:613. [PMID: 29258559 PMCID: PMC5738103 DOI: 10.1186/s13071-017-2575-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 12/06/2017] [Indexed: 11/16/2022] Open
Abstract
Background It is well known that reactive oxygen species (ROS) and reactive nitrogen species (RNS) are involved in the control of pathogens and microbiota in insects. However, the knowledge of the role of ROS and RNS in tick-pathogen and tick-microbiota interactions is limited. Here, we evaluated the immune-related redox metabolism of the embryonic cell line BME26 from the cattle tick Rhipicephalus microplus in response to Anaplasma marginale infection. Methods A high-throughput qPCR approach was used to determine the expression profile of 16 genes encoding proteins involved in either production or detoxification of ROS and RNS in response to different microbial challenges. In addition, the effect of RNAi-mediated gene silencing of catalase, glutathione peroxidase, thioredoxin and protein oxidation resistance 1 in the control of infection with A. marginale was evaluated. Results Infection with A. marginale resulted in downregulation of the genes encoding ROS-generating enzymes dual oxidase and endoplasmic reticulum oxidase. In contrast, the genes encoding the antioxidant enzymes superoxide dismutase, catalase, glutathione peroxidase, glutathione S-transferase, thioredoxin, thioredoxin reductase and peroxiredoxin were upregulated. The gene expression pattern in response to infection with Rickettsia rickettsii and exposure to heat-killed microorganisms, Micrococcus luteus, Enterobacter cloacae or S. cerevisiae was the opposite of that triggered by A. marginale challenge. The simultaneous silencing of three genes, catalase, glutathione peroxidase, and thioredoxin as well as the oxidation resistance 1 gene by RNAi apparently favoured the colonization of BME26 cells by A. marginale, suggesting that the antioxidant response might play a role in the control of infection. Conclusions Taken together, our results suggest that a general response of tick cells upon microbial stimuli is to increase ROS/RNS production. In contrast, A. marginale infection triggers an opposite profile, suggesting that this pathogen might manipulate the tick redox metabolism to evade the deleterious effect of the oxidant-based innate immune response. Electronic supplementary material The online version of this article (10.1186/s13071-017-2575-9) contains supplementary material, which is available to authorized users.
Collapse
|
39
|
Capelli-Peixoto J, Carvalho DD, Johnson WC, Scoles GA, Fogaça AC, Daffre S, Ueti MW. The transcription factor Relish controls Anaplasma marginale infection in the bovine tick Rhipicephalus microplus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 74:32-39. [PMID: 28408334 DOI: 10.1016/j.dci.2017.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/06/2017] [Accepted: 04/08/2017] [Indexed: 06/07/2023]
Abstract
Rhipicephalus microplus is an important biological vector of Anaplasma marginale, the etiological agent of bovine anaplasmosis. The knowledge of tick immune responses to control bacterial infections remains limited. In this study, we demonstrate that transcription factor Relish from the IMD signaling pathway has an important role in the control of A. marginale infection in ticks. We found that RNA-mediated silencing of Relish caused a significant increase in the number of A. marginale in the midgut and salivary glands of R. microplus. In addition, the IMD pathway regulates the expression of the gene that encodes the antimicrobial peptide (AMP) microplusin. Moreover, microplusin expression was up-regulated in the midgut (2×) and salivary glands (8×) of A. marginale infected R. microplus. Therefore, it is plausible to hypothesize that microplusin may be involved in the A. marginale control. This study provides the first evidence of IMD signaling pathway participation on the A. marginale control in R. microplus.
Collapse
Affiliation(s)
- J Capelli-Peixoto
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, State of São Paulo, Brazil.
| | - Danielle D Carvalho
- Department of Special Analysis, SD&W Modelagem e Soluções Estratégicas Ltda., 04707-010, São Paulo, State of São Paulo, Brazil.
| | - Wendell C Johnson
- Animal Diseases Research Unit, United States Department of Agricultural - Agricultural Research Service, Pullman, WA, United States.
| | - Glen A Scoles
- Animal Diseases Research Unit, United States Department of Agricultural - Agricultural Research Service, Pullman, WA, United States.
| | - Andrea C Fogaça
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, State of São Paulo, Brazil.
| | - Sirlei Daffre
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, State of São Paulo, Brazil.
| | - Massaro W Ueti
- Animal Diseases Research Unit, United States Department of Agricultural - Agricultural Research Service, Pullman, WA, United States.
| |
Collapse
|
40
|
Martins LA, Galletti MFBDM, Ribeiro JM, Fujita A, Costa FB, Labruna MB, Daffre S, Fogaça AC. The Distinct Transcriptional Response of the Midgut of Amblyomma sculptum and Amblyomma aureolatum Ticks to Rickettsia rickettsii Correlates to Their Differences in Susceptibility to Infection. Front Cell Infect Microbiol 2017; 7:129. [PMID: 28503490 PMCID: PMC5409265 DOI: 10.3389/fcimb.2017.00129] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 03/29/2017] [Indexed: 12/19/2022] Open
Abstract
Rickettsia rickettsii is a tick-borne obligate intracellular bacterium that causes Rocky Mountain Spotted Fever (RMSF). In Brazil, two species of ticks in the genus Amblyomma, A. sculptum and A. aureolatum, are incriminated as vectors of this bacterium. Importantly, these two species present remarkable differences in susceptibility to R. rickettsii infection, where A. aureolatum is more susceptible than A. sculptum. In the current study, A. aureolatum and A. sculptum ticks were fed on suitable hosts previously inoculated with R. rickettsii, mimicking a natural infection. As control, ticks were fed on non-infected animals. Both midgut and salivary glands of all positively infected ticks were colonized by R. rickettsii. We did not observe ticks with infection restricted to midgut, suggesting that important factors for controlling rickettsial colonization were produced in this organ. In order to identify such factors, the total RNA extracted from the midgut (MG) was submitted to next generation RNA sequencing (RNA-seq). The majority of the coding sequences (CDSs) of A. sculptum differentially expressed by infection were upregulated, whereas most of modulated CDSs of A. aureolatum were downregulated. The functional categories that comprise upregulated CDSs of A. sculptum, for instance, metabolism, signal transduction, protein modification, extracellular matrix, and immunity also include CDSs of A. aureolatum that were downregulated by infection. This is the first study that reports the effects of an experimental infection with the highly virulent R. rickettsii on the gene expression of two natural tick vectors. The distinct transcriptional profiles of MG of A. sculptum and A. aureolatum upon infection stimulus strongly suggest that molecular factors in this organ are responsible for delineating the susceptibility to R. rickettsii. Functional studies to determine the role played by proteins encoded by differentially expressed CDSs in the acquisition of R. rickettsii are warranted and may be considered as targets for the development of strategies to control the tick-borne pathogens as well as to control the tick vectors.
Collapse
Affiliation(s)
- Larissa A Martins
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São PauloSão Paulo, Brazil
| | - Maria F B de Melo Galletti
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São PauloSão Paulo, Brazil
| | - José M Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious DiseasesRockville, MD, USA
| | - André Fujita
- Departamento de Ciência da Computação, Instituto de Matemática e Estatística, Universidade de São PauloSão Paulo, Brazil
| | - Francisco B Costa
- Departamento de Medicina Veterinária Preventiva e Saúde Animal, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São PauloSão Paulo, Brazil
| | - Marcelo B Labruna
- Departamento de Medicina Veterinária Preventiva e Saúde Animal, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São PauloSão Paulo, Brazil
| | - Sirlei Daffre
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São PauloSão Paulo, Brazil
| | - Andréa C Fogaça
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São PauloSão Paulo, Brazil
| |
Collapse
|
41
|
Infection-derived lipids elicit an immune deficiency circuit in arthropods. Nat Commun 2017; 8:14401. [PMID: 28195158 PMCID: PMC5316886 DOI: 10.1038/ncomms14401] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 12/22/2016] [Indexed: 12/13/2022] Open
Abstract
The insect immune deficiency (IMD) pathway resembles the tumour necrosis factor receptor network in mammals and senses diaminopimelic-type peptidoglycans present in Gram-negative bacteria. Whether unidentified chemical moieties activate the IMD signalling cascade remains unknown. Here, we show that infection-derived lipids 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphoglycerol (POPG) and 1-palmitoyl-2-oleoyl diacylglycerol (PODAG) stimulate the IMD pathway of ticks. The tick IMD network protects against colonization by three distinct bacteria, that is the Lyme disease spirochete Borrelia burgdorferi and the rickettsial agents Anaplasma phagocytophilum and A. marginale. Cell signalling ensues in the absence of transmembrane peptidoglycan recognition proteins and the adaptor molecules Fas-associated protein with a death domain (FADD) and IMD. Conversely, biochemical interactions occur between x-linked inhibitor of apoptosis protein (XIAP), an E3 ubiquitin ligase, and the E2 conjugating enzyme Bendless. We propose the existence of two functionally distinct IMD networks, one in insects and another in ticks. The insect IMD signalling pathway detects invading pathogens. Here the authors show that ticks have an alternative IMD system that lacks peptidoglycan receptors, IMD and FADD, and is instead reliant on interaction of the E3 ligase XIAP with the E2 conjugating enzyme Bendless.
Collapse
|
42
|
Oliva Chávez AS, Shaw DK, Munderloh UG, Pedra JHF. Tick Humoral Responses: Marching to the Beat of a Different Drummer. Front Microbiol 2017; 8:223. [PMID: 28261180 PMCID: PMC5306392 DOI: 10.3389/fmicb.2017.00223] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/31/2017] [Indexed: 12/20/2022] Open
Abstract
Ticks transmit a variety of human pathogens, including Borrelia burgdorferi, the etiological agent of Lyme disease. Multiple pathogens that are transmitted simultaneously, termed “coinfections,” are of increasing importance and can affect disease outcome in a host. Arthropod immunity is central to pathogen acquisition and transmission by the tick. Pattern recognition receptors recognize pathogen-associated molecular patterns and induce humoral responses through the Toll and Immune Deficiency (IMD) pathways. Comparative analyses between insects and ticks reveal that while the Toll pathway is conserved, the IMD network exhibits a high degree of variability. This indicates that major differences in humoral immunity exist between insects and ticks. While many variables can affect immunity, one of the major forces that shape immune outcomes is the microbiota. In light of this, we discuss how the presence of commensal bacteria, symbionts and/or coinfections can lead to altered immune responses in the tick that impact pathogen persistence and subsequent transmission. By investigating non-insect arthropod immunity, we will not only better comprehend tick biology, but also unravel the intricate effects that pathogen coinfections have on vector competence and tick-borne disease transmission.
Collapse
Affiliation(s)
- Adela S Oliva Chávez
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore MD, USA
| | - Dana K Shaw
- 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
|
43
|
Kim BM, Kang S, Ahn DH, Kim JH, Ahn I, Lee CW, Cho JL, Min GS, Park H. First Insights into the Subterranean Crustacean Bathynellacea Transcriptome: Transcriptionally Reduced Opsin Repertoire and Evidence of Conserved Homeostasis Regulatory Mechanisms. PLoS One 2017; 12:e0170424. [PMID: 28107438 PMCID: PMC5249073 DOI: 10.1371/journal.pone.0170424] [Citation(s) in RCA: 8] [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: 09/09/2016] [Accepted: 01/04/2017] [Indexed: 11/25/2022] Open
Abstract
Bathynellacea (Crustacea, Syncarida, Parabathynellidae) are subterranean aquatic crustaceans that typically inhabit freshwater interstitial spaces (e.g., groundwater) and are occasionally found in caves and even hot springs. In this study, we sequenced the whole transcriptome of Allobathynella bangokensis using RNA-seq. De novo sequence assembly produced 74,866 contigs including 28,934 BLAST hits. Overall, the gene sequences were most similar to those of the waterflea Daphnia pulex. In the A. bangokensis transcriptome, no opsin or related sequences were identified, and no contig aligned to the crustacean visual opsins and non-visual opsins (i.e. arthropsins, peropsins, and melaopsins), suggesting potential regressive adaptation to the dark environment. However, A. bangokensis expressed conserved gene family sets, such as heat shock proteins and those related to key innate immunity pathways and antioxidant defense systems, at the transcriptional level, suggesting that this species has evolved adaptations involving molecular mechanisms of homeostasis. The transcriptomic information of A. bangokensis will be useful for investigating molecular adaptations and response mechanisms to subterranean environmental conditions.
Collapse
Affiliation(s)
- Bo-Mi Kim
- Unit of Polar Genomics, Korea Polar Research Institute, Incheon, South Korea
| | - Seunghyun Kang
- Unit of Polar Genomics, Korea Polar Research Institute, Incheon, South Korea
| | - Do-Hwan Ahn
- Unit of Polar Genomics, Korea Polar Research Institute, Incheon, South Korea
| | - Jin-Hyoung Kim
- Unit of Polar Genomics, Korea Polar Research Institute, Incheon, South Korea
| | - Inhye Ahn
- Unit of Polar Genomics, Korea Polar Research Institute, Incheon, South Korea
- Polar Sciences, University of Science & Technology, Yuseong-gu, Daejeon, South Korea
| | - Chi-Woo Lee
- Department of Biological Sciences, Inha University, Incheon, South Korea
| | - Joo-Lae Cho
- Nakdonggang National Institute of Biological Resources, Sangju, South Korea
| | - Gi-Sik Min
- Department of Biological Sciences, Inha University, Incheon, South Korea
| | - Hyun Park
- Unit of Polar Genomics, Korea Polar Research Institute, Incheon, South Korea
- Polar Sciences, University of Science & Technology, Yuseong-gu, Daejeon, South Korea
| |
Collapse
|