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
|
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
|
3
|
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
|
4
|
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
|
5
|
Zhu JJ, Zhang HZ, Hong RD, Yu D, Hong M, Liu ZX, Li DM, Yin JX. Prevalence and genetic diversity of Anaplasma phagocytophilum in wild small mammals from western Yunnan province, China. Front Vet Sci 2024; 11:1472595. [PMID: 39539313 PMCID: PMC11557534 DOI: 10.3389/fvets.2024.1472595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Anaplasma phagocytophilum (A. phagocytophilum) is an emerging zoonotic pathogen causing human granulocytic anaplasmosis, linked to small mammal reservoirs that harbor various zoonotic pathogens, underscoring their importance in public health and ecology. This study seeks to determine the prevalence of A. phagocytophilum in small mammals using PCR, then sequence and genotype positive samples, and assess infection risk factors. Small mammals were seasonally captured and a nested polymerase chain reaction (nested-PCR) was conducted targeting the 16S rRNA gene on spleen samples to detect A. phagocytophilum infection from three counties in western Yunnan province, China. Positive samples were sequenced and genotyped, revealing genetic diversity and regional clustering of the pathogen. A total of 1,605 small mammals belonging to 30 species, 18 genera, 6 families, 3 orders were captured seasonally and screened in this region, yielding a 0.93% infection rate with A. phagocytophilum (15/1605). Significant variations in infection rates were observed across different species, counties, and habitats. The 16Sr RNA genes of A. phagocytophilum were categorized into two distinct clades, indicating notable genetic diversity. The identification of genetic variants in spleen samples underscores the potential public health risk and the critical importance of the One Health approach in disease surveillance. Our findings emphasize the necessity for continuous monitoring and highlight the value of nested-PCR testing on spleen samples for accurate prevalence assessment.
Collapse
Affiliation(s)
- Jun-jie Zhu
- School of Public Health, Dali University, Dali, China
| | - Hong-ze Zhang
- School of Public Health, Dali University, Dali, China
| | - Ru-dan Hong
- School of Public Health, Dali University, Dali, China
| | - Dan Yu
- School of Public Health, Dali University, Dali, China
| | - Mei Hong
- Yunnan Institute of Endemic Disease Control and Prevention, Dali, China
| | - Zheng-xiang Liu
- Yunnan Institute of Endemic Disease Control and Prevention, Dali, China
| | - Dong-mei Li
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jia-xiang Yin
- School of Public Health, Dali University, Dali, China
| |
Collapse
|
6
|
Gęgotek A, Moniuszko-Malinowska A, Groth M, Skrzydlewska E. Changes in cerebrospinal fluid proteome of patients with tick-borne encephalitis. J Med Virol 2024; 96:e29763. [PMID: 38949193 DOI: 10.1002/jmv.29763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/11/2024] [Accepted: 06/11/2024] [Indexed: 07/02/2024]
Abstract
Tick-borne encephalitis (TBE) is one of the main diseases transmitted by ticks, the incidence of which is increasing. Moreover, its diagnosis and therapy are often long and difficult according to nonspecific symptoms and complex etiology. This study aimed to observe changes in the proteome of cerebrospinal fluid from TBE patients. Cerebrospinal fluid (CSF) of TBE patients (n = 20) and healthy individuals (n = 10) was analyzed using a proteomic approach (QExactiveHF-Orbitrap mass spectrometer) and zymography. Obtained results show that in CSF of TBE patients, the top-upregulated proteins are involved in pro-inflammatory reaction (interleukins), as well as antioxidant/protective response (peroxiredoxins, heat shock proteins). Moreover, changes in the proteome of CSF are not only the result of this disease development, but they can also be an indicator of its course. This mainly applies to proteins involved in proteolysis including serpins and metalloproteinases, whose activity is proportional to the length of patients' convalescence. The obtained proteomic data strongly direct attention to the changes caused by the development of TBE to antioxidant, pro-inflammatory, and proteolytic proteins, knowledge about which can significantly contribute to faster and more accurate diagnosis of various clinical forms of TBE.
Collapse
Affiliation(s)
- Agnieszka Gęgotek
- Department of Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| | - Anna Moniuszko-Malinowska
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland
| | - Monika Groth
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Bialystok, Poland
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
7
|
Reis AADL, de Avelar BR, Rocha MBDS, Borges DA, Campos DR, Fiorotti J, Golo PS, Scott FB. Ultrastructural characterization and quantification of hemocytes in engorged female Amblyomma sculptum ticks. Ticks Tick Borne Dis 2024; 15:102312. [PMID: 38277717 DOI: 10.1016/j.ttbdis.2024.102312] [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: 08/29/2023] [Revised: 12/26/2023] [Accepted: 01/03/2024] [Indexed: 01/28/2024]
Abstract
Amblyomma sculptum (formerly Amblyomma cajennense) ticks have been implicated in the transmission of pathogens that cause diseases in animals and humans. Their wide geographic distribution and high impact on animal health and zoonotic disease transmission highlight the importance of studying and implementing effective control measures to mitigate the risks associated with this tick species. The aim of this study was to quantify and characterize the morphology and the ultrastructure of different types of hemocytes in the hemolymph in engorged A. sculptum females fed on rabbits. The hemolymph samples were collected by perforation of the cuticle in the dorsal region. Hemocyte types, sizes, and differential counts were determined using light microscopy, while ultrastructural analysis of hemocytes was performed using transmission electron microscopy. The average number of total hemocytes in the hemolymph was 1024 ± 597.6 cells µL-1. Five morphologically distinct cell types were identified in A. sculptum females: prohemocytes (6 % ± 8.8), plasmatocytes (10 % ± 7.7), granulocytes (78 % ± 12.2), spherulocytes (5 % ± 4.48), and oenocytoids (1 % ± 1.6). In general, prohemocytes were the smallest hemocytes. The ultrastructural morphology of A. sculptum hemocytes described in the present study agrees with the findings for other hard ticks. This is the first study to investigate ultrastructural characteristics of hemocytes of female A. sculptum ticks.
Collapse
Affiliation(s)
- Andressa Aparecida de Lima Reis
- Department of Animal Parasitology, Veterinary Institute, Federal Rural University of Rio de Janeiro, Km 07, Zona Rural, BR-465, Seropédica, RJ 23890-000, Brazil.
| | - Barbara Rauta de Avelar
- Department of Animal Parasitology, Veterinary Institute, Federal Rural University of Rio de Janeiro, Km 07, Zona Rural, BR-465, Seropédica, RJ 23890-000, Brazil
| | - Marisa Beatriz da Silva Rocha
- Department of Animal Parasitology, Veterinary Institute, Federal Rural University of Rio de Janeiro, Km 07, Zona Rural, BR-465, Seropédica, RJ 23890-000, Brazil
| | - Debora Azevedo Borges
- Department of Animal Parasitology, Veterinary Institute, Federal Rural University of Rio de Janeiro, Km 07, Zona Rural, BR-465, Seropédica, RJ 23890-000, Brazil
| | - Diefrey Ribeiro Campos
- Department of Animal Parasitology, Veterinary Institute, Federal Rural University of Rio de Janeiro, Km 07, Zona Rural, BR-465, Seropédica, RJ 23890-000, Brazil
| | - Jessica Fiorotti
- Department of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo 14040-900, Brazil
| | - Patrícia Silva Golo
- Department of Animal Parasitology, Veterinary Institute, Federal Rural University of Rio de Janeiro, Km 07, Zona Rural, BR-465, Seropédica, RJ 23890-000, Brazil
| | - Fabio Barbour Scott
- Department of Animal Parasitology, Veterinary Institute, Federal Rural University of Rio de Janeiro, Km 07, Zona Rural, BR-465, Seropédica, RJ 23890-000, Brazil
| |
Collapse
|
8
|
Zhao C, Cai G, Zhang X, Liu X, Wang P, Zheng A. Comparative Analysis of Bisexual and Parthenogenetic Populations in Haemaphysalis Longicornis. Microorganisms 2024; 12:823. [PMID: 38674766 PMCID: PMC11051975 DOI: 10.3390/microorganisms12040823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Haemaphysalis longicornis, a three-host tick with a wide host range, is widely distributed in different countries and regions. It stands out among ticks due to its unique feature of having both parthenogenetic and bisexual populations. Despite their morphological resemblance, the characteristics of the parthenogenetic population have been overlooked. In this comprehensive study, we systematically compared the similarities and differences between these two populations. Our investigation revealed that the parthenogenetic H. longicornis, widely distributed in China, was found in ten provinces, surpassing the previously reported distribution. Notably, individuals from the parthenogenetic population exhibited a prolonged blood-feeding duration during the larval and nymph stages compared to their bisexual counterparts. Additionally, the life cycle of the parthenogenetic population was observed to be longer. A flow cytometry analysis indicated a DNA content ratio of approximately 2:3 between the bisexual and parthenogenetic populations. A phylogenetic analysis using whole mitochondrial genome sequences resulted in the separation of the phylogenetic tree into two distinct branches. A molecular analysis unveiled a consistent single T-base deletion at nucleotide 8497 in the parthenogenetic population compared to the bisexual population. Both populations displayed high viral infection capability and significant resistance to ivermectin. Intriguingly, despite these differences, the parthenogenetic population exhibited a similar life cycle to the bisexual population, retaining the ability to transmit pathogens such as Severe fever with thrombocytopenia syndrome virus (SFTSV) and Heartland Virus (HRTV). These findings contribute to a deeper understanding of the distinct characteristics and similarities between different populations of H. longicornis, laying the foundation for future research in this field.
Collapse
Affiliation(s)
- Chaoyue Zhao
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, School of Life Sciences, Fudan University, Shanghai 200437, China; (C.Z.); (G.C.); (X.L.)
| | - Guonan Cai
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, School of Life Sciences, Fudan University, Shanghai 200437, China; (C.Z.); (G.C.); (X.L.)
| | - Xing Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China;
| | - Xinyu Liu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, School of Life Sciences, Fudan University, Shanghai 200437, China; (C.Z.); (G.C.); (X.L.)
- Aulin College, Northeast Forestry University, Harbin 150040, China
| | - Pengfei Wang
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, School of Life Sciences, Fudan University, Shanghai 200437, China; (C.Z.); (G.C.); (X.L.)
| | - Aihua Zheng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China;
| |
Collapse
|
9
|
Wang Y, Xu Z, Zhang H, Zhou Y, Cao J, Zhang Y, Wang Z, Zhou J. Towards modelling tick-virus interactions using the weakly pathogenic Sindbis virus: Evidence that ticks are competent vectors. Front Cell Infect Microbiol 2024; 14:1334351. [PMID: 38567020 PMCID: PMC10985168 DOI: 10.3389/fcimb.2024.1334351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/27/2024] [Indexed: 04/04/2024] Open
Abstract
Most tick-borne viruses (TBVs) are highly pathogenic and require high biosecurity, which severely limits their study. We found that Sindbis virus (SINV), predominantly transmitted by mosquitoes, can replicate in ticks and be subsequently transmitted, with the potential to serve as a model for studying tick-virus interactions. We found that both larval and nymphal stages of Rhipicephalus haemaphysaloides can be infected with SINV-wild-type (WT) when feeding on infected mice. SINV replicated in two species of ticks (R. haemaphysaloides and Hyalomma asiaticum) after infecting them by microinjection. Injection of ticks with SINV expressing enhanced Green Fluorescent Protein (eGFP) revealed that SINV-eGFP specifically aggregated in the tick midguts for replication. During blood-feeding, SINV-eGFP migrated from the midguts to the salivary glands and was transmitted to a new host. SINV infection caused changes in expression levels of tick genes related to immune responses, substance transport and metabolism, cell growth and death. SINV mainly induced autophagy during the early stage of infection; with increasing time of infection, the level of autophagy decreased, while the level of apoptosis increased. During the early stages of infection, the transcript levels of immune-related genes were significantly upregulated, and then decreased. In addition, SINV induced changes in the transcription levels of some functional genes that play important roles in the interactions between ticks and tick-borne pathogens. These results confirm that the SINV-based transmission model between ticks, viruses, and mammals can be widely used to unravel the interactions between ticks and viruses.
Collapse
Affiliation(s)
- Yanan Wang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Zhengmao Xu
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Houshuang Zhang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yongzhi Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Jie Cao
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yuqiang Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Zedong Wang
- Center of Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Jilin, China
| | - Jinlin Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| |
Collapse
|
10
|
Deng YP, Fu YT, Yao C, Shao R, Zhang XL, Duan DY, Liu GH. Emerging bacterial infectious diseases/pathogens vectored by human lice. Travel Med Infect Dis 2023; 55:102630. [PMID: 37567429 DOI: 10.1016/j.tmaid.2023.102630] [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: 05/12/2023] [Revised: 07/02/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Human lice have always been a major public health concern due to their vector capacity for louse-borne infectious diseases, like trench fever, louse-borne relapsing fever, and epidemic fever, which are caused by Bartonella quintana, Borrelia recurrentis, and Rickettsia prowazekii, respectively. Those diseases are currently re-emerging in the regions of poor hygiene, social poverty, or wars with life-threatening consequences. These louse-borne diseases have also caused outbreaks among populations in jails and refugee camps. In addition, antibodies and DNAs to those pathogens have been steadily detected in homeless populations. Importantly, more bacterial pathogens have been detected in human lice, and some have been transmitted by human lice in laboratories. Here, we provide a comprehensive review and update on louse-borne infectious diseases/bacterial pathogens.
Collapse
Affiliation(s)
- Yuan-Ping Deng
- Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan, China
| | - Yi-Tian Fu
- Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan, China; Department of Parasitology, Xiangya School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Chaoqun Yao
- Department of Biomedical Sciences and One Health Center for Zoonoses and Tropical Veterinary Medicine, Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis.
| | - Renfu Shao
- Centre for Bioinnovation, School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, Australia
| | - Xue-Ling Zhang
- Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan, China
| | - De-Yong Duan
- Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan, China
| | - Guo-Hua Liu
- Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha, Hunan, China.
| |
Collapse
|
11
|
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
|
12
|
Yousefi-Behzadi M, Mehrabi A, Ahmadinezhad M, Rohani M, Naddaf SR, Bagheri A, Shams-Ghahfarokhi M, Maghsoudlou E, Mojahed N, Mounesan L, Tahmasebi Z, Sohrabi A, Salehi-Vaziri M, Salehi Z, Razzaghi-Abyaneh M. Metagenomics Characterization of Ixodes ricinus Intestinal Microbiota as Major Vector of Tick-Borne Diseases in Domestic Animals. J Arthropod Borne Dis 2023; 17:152-164. [PMID: 37822757 PMCID: PMC10562206 DOI: 10.18502/jad.v17i2.13620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 06/11/2023] [Indexed: 10/13/2023] Open
Abstract
Background Understanding the microbiota of disease vectors can help for developing new strategies to prevent the transmission of vector pathogens. Ixodes ricinus is one of the most notorious tick vectors with increasing importance in Iran and other parts of the world while there is limited data on its microbiota. This study aimed to use metagenomics for identifying the I. ricinus tick's microbiota of Iran. Methods A total of 39 adult ticks were collected from Mazandaran (21 females), Gilan (17 females), and Golestan (1 male). Five tick pools prepared from 39 adults of I. ricinus were subjected to metagenomics analysis. The data were analyzed by targeting the V6 region of the 16S rRNA gene by Illumina 4000 Hiseq sequencing. Results Among hundreds of intestinal microbiota identified by metagenomics, various pathogenic microorganisms distributed in 30 genera and species including those responsible for tick-borne diseases resided in the genera Coxiella, Rickettsia, and Burkholderia were found. Conclusion Our results indicated that metagenomics identifies bacteria genera and species which cannot be easily recognized by routine methods. The presence of such pathogenic bacteria indicates the importance of possible zoonotic diseases in this region which could affect public health. These results further substantiate the importance of advanced metagenomics analyses to identify neglected tick-borne pathogens which enable researchers to provide efficient mapping roads for the management of emerging and re-emerging infectious diseases.
Collapse
Affiliation(s)
- Manijeh Yousefi-Behzadi
- Department of Epidemiology and Biostatics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
- National Reference Laboratory of Plague, Tularemia and Q Fever, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Akanlu, Kabudar-Ahang, Hamadan, Iran
| | - Atefeh Mehrabi
- School of Advanced Medical Sciences, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Mozhgan Ahmadinezhad
- Department of Epidemiology and Biostatics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Rohani
- Department of Microbiology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Amin Bagheri
- Department of Pathobiology, Faculty of Veterinary Medicine, Science and Research, Tehran, Iran
| | | | - Ebrahim Maghsoudlou
- Department of Computer Science, School of Computing, Southern Illinois University Carbondale, Illinois
| | - Nooshin Mojahed
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, Illinois
| | - Leila Mounesan
- Department of Epidemiology and Biostatics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Zahra Tahmasebi
- Department of Epidemiology and Biostatics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
- National Reference Laboratory of Plague, Tularemia and Q Fever, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Akanlu, Kabudar-Ahang, Hamadan, Iran
| | - Aria Sohrabi
- Department of Epidemiology and Biostatics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
- National Reference Laboratory of Plague, Tularemia and Q Fever, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Akanlu, Kabudar-Ahang, Hamadan, Iran
| | - Mostafa Salehi-Vaziri
- Department of Arboviruses and Viral Hemorrhagic Fevers, Pasteur Institute of Iran, Tehran, Iran
| | - Zahra Salehi
- Department of Mycology, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
13
|
Feng C, Torimaru K, Lim MYT, Chak LL, Shiimori M, Tsuji K, Tanaka T, Iida J, Okamura K. A novel eukaryotic RdRP-dependent small RNA pathway represses antiviral immunity by controlling an ERK pathway component in the black-legged tick. PLoS One 2023; 18:e0281195. [PMID: 36996253 PMCID: PMC10062562 DOI: 10.1371/journal.pone.0281195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 01/17/2023] [Indexed: 04/01/2023] Open
Abstract
Small regulatory RNAs (sRNAs) are involved in antiviral defense and gene regulation. Although roles of RNA-dependent RNA Polymerases (RdRPs) in sRNA biology are extensively studied in nematodes, plants and fungi, understanding of RdRP homologs in other animals is still lacking. Here, we study sRNAs in the ISE6 cell line, which is derived from the black-legged tick, an important vector of human and animal pathogens. We find abundant classes of ~22nt sRNAs that require specific combinations of RdRPs and sRNA effector proteins (Argonautes or AGOs). RdRP1-dependent sRNAs possess 5'-monophosphates and are mainly derived from RNA polymerase III-transcribed genes and repetitive elements. Knockdown of some RdRP homologs misregulates genes including RNAi-related genes and the regulator of immune response Dsor1. Sensor assays demonstrate that Dsor1 is downregulated by RdRP1 through the 3'UTR that contains a target site of RdRP1-dependent repeat-derived sRNAs. Consistent with viral gene repression by the RNAi mechanism using virus-derived small interfering RNAs, viral transcripts are upregulated by AGO knockdown. On the other hand, RdRP1 knockdown unexpectedly results in downregulation of viral transcripts. This effect is dependent on Dsor1, suggesting that antiviral immunity is enhanced by RdRP1 knockdown through Dsor1 upregulation. We propose that tick sRNA pathways control multiple aspects of immune response via RNAi and regulation of signaling pathways.
Collapse
Affiliation(s)
- Canran Feng
- Nara Institute of Science and Technology, Nara, Japan
| | | | - Mandy Yu Theng Lim
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Li-Ling Chak
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, Singapore
| | | | - Kosuke Tsuji
- Nara Institute of Science and Technology, Nara, Japan
| | - Tetsuya Tanaka
- Joint Faculty of Veterinary Medicine, Laboratory of Infectious Diseases, Kagoshima University, Kagoshima, Japan
| | - Junko Iida
- Nara Institute of Science and Technology, Nara, Japan
| | - Katsutomo Okamura
- Nara Institute of Science and Technology, Nara, Japan
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
14
|
A systems biology approach to better understand human tick-borne diseases. Trends Parasitol 2023; 39:53-69. [PMID: 36400674 DOI: 10.1016/j.pt.2022.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 11/17/2022]
Abstract
Tick-borne diseases (TBDs) are a growing global health concern. Despite extensive studies, ill-defined tick-associated pathologies remain with unknown aetiologies. Human immunological responses after tick bite, and inter-individual variations of immune-response phenotypes, are not well characterised. Current reductive experimental methodologies limit our understanding of more complex tick-associated illness, which results from the interactions between the host, tick, and microbes. An unbiased, systems-level integration of clinical metadata and biological host data - obtained via transcriptomics, proteomics, and metabolomics - offers to drive the data-informed generation of testable hypotheses in TBDs. Advanced computational tools have rendered meaningful analysis of such large data sets feasible. This review highlights the advantages of integrative system biology approaches as essential for understanding the complex pathobiology of TBDs.
Collapse
|
15
|
Hodosi R, Kazimirova M, Soltys K. What do we know about the microbiome of I. ricinus? Front Cell Infect Microbiol 2022; 12:990889. [PMID: 36467722 PMCID: PMC9709289 DOI: 10.3389/fcimb.2022.990889] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 10/17/2022] [Indexed: 10/07/2023] Open
Abstract
I. ricinus is an obligate hematophagous parasitic arthropod that is responsible for the transmission of a wide range of zoonotic pathogens including spirochetes of the genus Borrelia, Rickettsia spp., C. burnetii, Anaplasma phagocytophilum and Francisella tularensis, which are part the tick´s microbiome. Most of the studies focus on "pathogens" and only very few elucidate the role of "non-pathogenic" symbiotic microorganisms in I. ricinus. While most of the members of the microbiome are leading an intracellular lifestyle, they are able to complement tick´s nutrition and stress response having a great impact on tick´s survival and transmission of pathogens. The composition of the tick´s microbiome is not consistent and can be tied to the environment, tick species, developmental stage, or specific organ or tissue. Ovarian tissue harbors a stable microbiome consisting mainly but not exclusively of endosymbiotic bacteria, while the microbiome of the digestive system is rather unstable, and together with salivary glands, is mostly comprised of pathogens. The most prevalent endosymbionts found in ticks are Rickettsia spp., Ricketsiella spp., Coxiella-like and Francisella-like endosymbionts, Spiroplasma spp. and Candidatus Midichloria spp. Since microorganisms can modify ticks' behavior, such as mobility, feeding or saliva production, which results in increased survival rates, we aimed to elucidate the potential, tight relationship, and interaction between bacteria of the I. ricinus microbiome. Here we show that endosymbionts including Coxiella-like spp., can provide I. ricinus with different types of vitamin B (B2, B6, B7, B9) essential for eukaryotic organisms. Furthermore, we hypothesize that survival of Wolbachia spp., or the bacterial pathogen A. phagocytophilum can be supported by the tick itself since coinfection with symbiotic Spiroplasma ixodetis provides I. ricinus with complete metabolic pathway of folate biosynthesis necessary for DNA synthesis and cell division. Manipulation of tick´s endosymbiotic microbiome could present a perspective way of I. ricinus control and regulation of spread of emerging bacterial pathogens.
Collapse
Affiliation(s)
- Richard Hodosi
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
| | - Maria Kazimirova
- Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Katarina Soltys
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
- Comenius University Science Park, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
16
|
Wen S, Xu X, Kong J, Luo L, Yue P, Cao W, Zhang Y, Liu M, Fan Y, Chen J, Ma M, Tao L, Peng Y, Wang F, Dong Y, Li B, Luo S, Zhou G, Chen T, Li L, Liu A, Bao F. Comprehensive analyses of transcriptomes induced by Lyme spirochete infection to CNS model system. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2022; 103:105349. [PMID: 35964914 DOI: 10.1016/j.meegid.2022.105349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 06/12/2022] [Accepted: 08/09/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Lyme disease is a zoonotic disease caused by infection with Borrelia burgdorferi (Bb), the involvement of the nervous system in Lyme disease is usually referred to as Lyme neuroborreliosis (LNB). LNB has diverse clinical manifestations, most commonly including meningitis, Bell's palsy, and encephalitis. However, the molecular pathogenesis of neuroborreliosis is still poorly understood. Comprehensive transcriptomic analysis following Bb infection could provide new insights into the pathogenesis of LNB and may identify novel biomarkers or therapeutic targets for LNB diagnosis and treatment. METHODS In the present study, we pooled transcriptomic dataset of Macaca mulatta (rhesus) from our laboratory and the human astrocyte dataset GSE85143 from the Gene Expression Omnibus database to screen common differentially expressed genes (DEGs) in the Bb infection group and the control group. Functional and enrichment analyses were applied for the DEGs. Protein-Protein Interaction network, and hub genes were identified using the Search Tool for the Retrieval of Interaction Genes database and the CytoHubba plugin. Finally, mRNA expression of hub genes was validated in vitro and ex vivo from Bb infected models and normal controls by quantitative reverse transcription PCR (qRT-PCR). RESULTS A total of 80 upregulated DEGs and 32 downregulated DEGs were identified. Among them, 11 hub genes were selected. The pathway enrichment analyses on 11 hub genes revealed that the PI3K-Akt signaling pathway was significantly enriched. The mRNA levels of ANGPT1, TLR6, SREBF1, LDLR, TNC, and ITGA2 in U251 cells and/or rhesus brain explants by exposure to Bb were validated by qRT-PCR. CONCLUSION Our study suggested that TLR6, ANGPT1, LDLR, SREBF1, TNC, and ITGA may be candidate mammal biomarkers for LNB, and the TLR6/PI3K-Akt signaling pathway may play an important role in LNB pathogenesis.
Collapse
Affiliation(s)
- Shiyuan Wen
- Department of Microbiology and Immunology, Kunming Medical University, Kunming 650500, China; Department of Intensive Care Unit, First People's Hospital of Yunnan Province, Kunming 650500, China
| | - Xin Xu
- Department of Microbiology and Immunology, Kunming Medical University, Kunming 650500, China
| | - Jing Kong
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming 650500, China
| | - Lisha Luo
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming 650500, China
| | - Peng Yue
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming 650500, China
| | - Wenjing Cao
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming 650500, China
| | - Yu Zhang
- Department of Microbiology and Immunology, Kunming Medical University, Kunming 650500, China
| | - Meixiao Liu
- Department of Microbiology and Immunology, Kunming Medical University, Kunming 650500, China
| | - Yuxin Fan
- Department of Microbiology and Immunology, Kunming Medical University, Kunming 650500, China
| | - Jingjing Chen
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming 650500, China
| | - Mingbiao Ma
- Department of Microbiology and Immunology, Kunming Medical University, Kunming 650500, China; Yunnan Province Key Laboratory of Children's Major Diseases Research, The Affiliated Children's Hospital of Kunming, Kunming Medical University, Kunming 650030, China
| | - Lvyan Tao
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming 650500, China; Yunnan Province Key Laboratory of Children's Major Diseases Research, The Affiliated Children's Hospital of Kunming, Kunming Medical University, Kunming 650030, China
| | - Yun Peng
- Department of Microbiology and Immunology, Kunming Medical University, Kunming 650500, China
| | - Feng Wang
- Department of Microbiology and Immunology, Kunming Medical University, Kunming 650500, China
| | - Yan Dong
- Department of Microbiology and Immunology, Kunming Medical University, Kunming 650500, China
| | - Bingxue Li
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming 650500, China
| | - Suyi Luo
- Department of Microbiology and Immunology, Kunming Medical University, Kunming 650500, China
| | - Guozhong Zhou
- Department of Microbiology and Immunology, Kunming Medical University, Kunming 650500, China
| | - Taigui Chen
- Department of Microbiology and Immunology, Kunming Medical University, Kunming 650500, China
| | - Lianbao Li
- Department of Microbiology and Immunology, Kunming Medical University, Kunming 650500, China
| | - Aihua Liu
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming 650500, China; Yunnan Province Key Laboratory of Children's Major Diseases Research, The Affiliated Children's Hospital of Kunming, Kunming Medical University, Kunming 650030, China; The Institute for Tropical Medicine, Kunming Medical University, Kunming 650500, China; Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases, Kunming 650500, China.
| | - Fukai Bao
- Department of Microbiology and Immunology, Kunming Medical University, Kunming 650500, China; Yunnan Province Key Laboratory of Children's Major Diseases Research, The Affiliated Children's Hospital of Kunming, Kunming Medical University, Kunming 650030, China; The Institute for Tropical Medicine, Kunming Medical University, Kunming 650500, China; Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases, Kunming 650500, China.
| |
Collapse
|
17
|
Wang XR, Cull B. Apoptosis and Autophagy: Current Understanding in Tick–Pathogen Interactions. Front Cell Infect Microbiol 2022; 12:784430. [PMID: 35155277 PMCID: PMC8829008 DOI: 10.3389/fcimb.2022.784430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/11/2022] [Indexed: 11/13/2022] Open
Abstract
Tick-borne diseases are a significant threat to human and animal health throughout the world. How tick-borne pathogens successfully infect and disseminate in both their vertebrate and invertebrate hosts is only partially understood. Pathogens have evolved several mechanisms to combat host defense systems, and to avoid and modulate host immunity during infection, therefore benefitting their survival and replication. In the host, pathogens trigger responses from innate and adaptive immune systems that recognize and eliminate invaders. Two important innate defenses against pathogens are the programmed cell death pathways of apoptosis and autophagy. This Mini Review surveys the current knowledge of apoptosis and autophagy pathways in tick-pathogen interactions, as well as the strategies evolved by pathogens for their benefit. We then assess the limitations to studying both pathways and discuss their participation in the network of the tick immune system, before highlighting future perspectives in this field. The knowledge gained would significantly enhance our understanding of the defense responses in vector ticks that regulate pathogen infection and burden, and form the foundation for future research to identify novel approaches to the control of tick-borne diseases.
Collapse
Affiliation(s)
- Xin-Ru Wang
- *Correspondence: Xin-Ru Wang, ; Benjamin Cull,
| | | |
Collapse
|
18
|
Viglietta M, Bellone R, Blisnick AA, Failloux AB. Vector Specificity of Arbovirus Transmission. Front Microbiol 2021; 12:773211. [PMID: 34956136 PMCID: PMC8696169 DOI: 10.3389/fmicb.2021.773211] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/19/2021] [Indexed: 12/20/2022] Open
Abstract
More than 25% of human infectious diseases are vector-borne diseases (VBDs). These diseases, caused by pathogens shared between animals and humans, are a growing threat to global health with more than 2.5 million annual deaths. Mosquitoes and ticks are the main vectors of arboviruses including flaviviruses, which greatly affect humans. However, all tick or mosquito species are not able to transmit all viruses, suggesting important molecular mechanisms regulating viral infection, dissemination, and transmission by vectors. Despite the large distribution of arthropods (mosquitoes and ticks) and arboviruses, only a few pairings of arthropods (family, genus, and population) and viruses (family, genus, and genotype) successfully transmit. Here, we review the factors that might limit pathogen transmission: internal (vector genetics, immune responses, microbiome including insect-specific viruses, and coinfections) and external, either biotic (adult and larvae nutrition) or abiotic (temperature, chemicals, and altitude). This review will demonstrate the dynamic nature and complexity of virus–vector interactions to help in designing appropriate practices in surveillance and prevention to reduce VBD threats.
Collapse
Affiliation(s)
- Marine Viglietta
- Unit of Arboviruses and Insect Vectors, Institut Pasteur, Sorbonne Université, Paris, France
| | - Rachel Bellone
- Unit of Arboviruses and Insect Vectors, Institut Pasteur, Sorbonne Université, Paris, France
| | - Adrien Albert Blisnick
- Unit of Arboviruses and Insect Vectors, Institut Pasteur, Sorbonne Université, Paris, France
| | - Anna-Bella Failloux
- Unit of Arboviruses and Insect Vectors, Institut Pasteur, Sorbonne Université, Paris, France
| |
Collapse
|
19
|
Salata C, Moutailler S, Attoui H, Zweygarth E, Decker L, Bell-Sakyi L. How relevant are in vitro culture models for study of tick-pathogen interactions? Pathog Glob Health 2021; 115:437-455. [PMID: 34190676 PMCID: PMC8635668 DOI: 10.1080/20477724.2021.1944539] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Although tick-borne infectious diseases threaten human and animal health worldwide, with constantly increasing incidence, little knowledge is available regarding vector-pathogen interactions and pathogen transmission. In vivo laboratory study of these subjects using live, intact ticks is expensive, labor-intensive, and challenging from the points of view of biosafety and ethics. Several in vitro models have been developed, including over 70 continuous cell lines derived from multiple tick species and a variety of tick organ culture systems, facilitating many research activities. However, some limitations have to be considered in the translation of the results from the in vitro environment to the in vivo situation of live, intact ticks, and vertebrate hosts. In this review, we describe the available in vitro models and selected results from their application to the study of tick-borne viruses, bacteria, and protozoa, where possible comparing these results to studies in live, intact ticks. Finally, we highlight the strengths and weaknesses of in vitro tick culture models and their essential role in tick-borne pathogen research.
Collapse
Affiliation(s)
- Cristiano Salata
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Sara Moutailler
- Laboratoire De Santé Animale, Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Maisons-Alfort, France
| | - Houssam Attoui
- Department of Animal Health, UMR1161 Virologie, INRAE, Ecole Nationale Vétérinaire d’Alfort, ANSES, Université Paris-Est, Maisons-Alfort, France
| | - Erich Zweygarth
- The Department of Veterinary Tropical Diseases, University of Pretoria, Pretoria, South Africa
| | - Lygia Decker
- Department of Preventive Veterinary Medicine, School of Veterinary Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Lesley Bell-Sakyi
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
20
|
Luzzi MDC, Carvalho LALD, Pinheiro DG, Lima-Duarte L, Camargo JV, Kishi LT, Fernandes CC, Machado RZ, Soares JF, André MR, Barros-Battesti DM. Analysis on the prokaryotic microbiome in females and embryonic cell cultures of Rhipicephalus sanguineus tropical and temperate lineages from two specific localities in Brazil. ACTA ACUST UNITED AC 2021; 30:e005721. [PMID: 34378769 DOI: 10.1590/s1984-29612021066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/15/2021] [Indexed: 01/04/2023]
Abstract
Two lineages of Rhipicephalus sanguineus are known in Brazil: the temperate or southern and the tropical or northern populations. The distribution patterns of both lineages of R. sanguineus have epidemiological implications that can affect vectorial competence concerning Ehrlichia canis, the agent of canine monocytic ehrlichiosis. Intending to identify the microbiomes of both lineages and compare microorganisms in R. sanguineus, we used the 16S rRNA (V4-V5 region) gene-based metataxonomic approach, through NGS sequencing on the MiSeq Illumina platform. We selected specimens of females from the environment and samples of primary embryonic cell cultures, from both lineages, and this was the first study to investigate the prokaryotic microbiome in tick cell cultures. The results showed that many bacterial taxa detected in the samples were typical members of the host environment. A significant diversity of microorganisms in R. sanguineus females and in embryonic cell cultures from both lineages was found, with emphasis on the presence of Coxiella in all samples, albeit in different proportions. The Coxiella species present in the two lineages of ticks may be different and may have co-evolved with them, thus driving different patterns of interactions between ticks and the pathogens that they can harbor or transmit to vertebrate hosts.
Collapse
Affiliation(s)
- Mayara de Cassia Luzzi
- Departamento de Patologia, Reprodução e Saúde Única, Faculdade de Ciências Agrárias e Veterinárias - FCAV, Universidade Estadual Paulista Júlio de Mesquita Filho - UNESP, Jaboticabal, SP, Brasil
| | - Lucas Amoroso Lopes de Carvalho
- Departamento de Tecnologia, Faculdade de Ciências Agrárias e Veterinárias - FCAV, Universidade Estadual Paulista Júlio de Mesquita Filho - UNESP, Jaboticabal, SP, Brasil
| | - Daniel Guariz Pinheiro
- Departamento de Tecnologia, Faculdade de Ciências Agrárias e Veterinárias - FCAV, Universidade Estadual Paulista Júlio de Mesquita Filho - UNESP, Jaboticabal, SP, Brasil
| | - Leidiane Lima-Duarte
- Departamento de Medicina Veterinária Preventiva e Saúde Animal, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo - USP, SP, Brasil
| | - Jaqueline Valéria Camargo
- Departamento de Patologia, Reprodução e Saúde Única, Faculdade de Ciências Agrárias e Veterinárias - FCAV, Universidade Estadual Paulista Júlio de Mesquita Filho - UNESP, Jaboticabal, SP, Brasil
| | - Luciano Takeshi Kishi
- Laboratório Multiusuário Centralizado para Sequenciamento de DNA em Larga Escala e Análise de Expressão Gênica - LMSeq, Departamento de Tecnologia, Faculdade de Ciências Agrárias e Veterinárias - FCAV, Universidade Estadual Paulista Júlio de Mesquita Filho - UNESP, Jaboticabal, SP, Brasil
| | - Camila Cesário Fernandes
- Laboratório Multiusuário Centralizado para Sequenciamento de DNA em Larga Escala e Análise de Expressão Gênica - LMSeq, Departamento de Tecnologia, Faculdade de Ciências Agrárias e Veterinárias - FCAV, Universidade Estadual Paulista Júlio de Mesquita Filho - UNESP, Jaboticabal, SP, Brasil
| | - Rosangela Zacarias Machado
- Departamento de Patologia, Reprodução e Saúde Única, Faculdade de Ciências Agrárias e Veterinárias - FCAV, Universidade Estadual Paulista Júlio de Mesquita Filho - UNESP, Jaboticabal, SP, Brasil
| | - João Fábio Soares
- Universidade Federal do Rio Grande do Sul - UFRGS, Porto Alegre, RS, Brasil
| | - Marcos Rogério André
- Departamento de Patologia, Reprodução e Saúde Única, Faculdade de Ciências Agrárias e Veterinárias - FCAV, Universidade Estadual Paulista Júlio de Mesquita Filho - UNESP, Jaboticabal, SP, Brasil
| | - Darci Moraes Barros-Battesti
- Departamento de Patologia, Reprodução e Saúde Única, Faculdade de Ciências Agrárias e Veterinárias - FCAV, Universidade Estadual Paulista Júlio de Mesquita Filho - UNESP, Jaboticabal, SP, Brasil.,Departamento de Medicina Veterinária Preventiva e Saúde Animal, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo - USP, SP, Brasil
| |
Collapse
|
21
|
Pan Y, Cheng A, Wang M, Yin Z, Jia R. The Dual Regulation of Apoptosis by Flavivirus. Front Microbiol 2021; 12:654494. [PMID: 33841381 PMCID: PMC8024479 DOI: 10.3389/fmicb.2021.654494] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 02/25/2021] [Indexed: 12/11/2022] Open
Abstract
Apoptosis is a form of programmed cell death, which maintains cellular homeostasis by eliminating pathogen-infected cells. It contains three signaling pathways: death receptor pathway, mitochondria-mediated pathway, and endoplasmic reticulum pathway. Its importance in host defenses is highlighted by the observation that many viruses evade, hinder or destroy apoptosis, thereby weakening the host’s immune response. Flaviviruses such as Dengue virus, Japanese encephalitis virus, and West Nile virus utilize various strategies to activate or inhibit cell apoptosis. This article reviews the research progress of apoptosis mechanism during flaviviruses infection, including flaviviruses proteins and subgenomic flaviviral RNA to regulate apoptosis by interacting with host proteins, as well as various signaling pathways involved in flaviviruses-induced apoptosis, which provides a scientific basis for understanding the pathogenesis of flaviviruses and helps in developing an effective antiviral therapy.
Collapse
Affiliation(s)
- Yuhong Pan
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Anchun Cheng
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Mingshu Wang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Zhongqiong Yin
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Renyong Jia
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| |
Collapse
|
22
|
Lemasson M, Caignard G, Unterfinger Y, Attoui H, Bell-Sakyi L, Hirchaud E, Moutailler S, Johnson N, Vitour D, Richardson J, Lacour SA. Exploration of binary protein-protein interactions between tick-borne flaviviruses and Ixodes ricinus. Parasit Vectors 2021; 14:144. [PMID: 33676573 PMCID: PMC7937244 DOI: 10.1186/s13071-021-04651-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/18/2021] [Indexed: 12/23/2022] Open
Abstract
Background Louping ill virus (LIV) and tick-borne encephalitis virus (TBEV) are tick-borne flaviviruses that are both transmitted by the major European tick, Ixodes ricinus. Despite the importance of I. ricinus as an arthropod vector, its capacity to acquire and subsequently transmit viruses, known as vector competence, is poorly understood. At the molecular scale, vector competence is governed in part by binary interactions established between viral and cellular proteins within infected tick cells. Methods To investigate virus-vector protein–protein interactions (PPIs), the entire set of open reading frames for LIV and TBEV was screened against an I. ricinus cDNA library established from three embryonic tick cell lines using yeast two-hybrid methodology (Y2H). PPIs revealed for each viral bait were retested in yeast by applying a gap repair (GR) strategy, and notably against the cognate protein of both viruses, to determine whether the PPIs were specific for a single virus or common to both. The interacting tick proteins were identified by automatic BLASTX, and in silico analyses were performed to expose the biological processes targeted by LIV and TBEV. Results For each virus, we identified 24 different PPIs involving six viral proteins and 22 unique tick proteins, with all PPIs being common to both viruses. According to our data, several viral proteins (pM, M, NS2A, NS4A, 2K and NS5) target multiple tick protein modules implicated in critical biological pathways. Of note, the NS5 and pM viral proteins establish PPI with several tumor necrosis factor (TNF) receptor-associated factor (TRAF) proteins, which are essential adaptor proteins at the nexus of multiple signal transduction pathways. Conclusion We provide the first description of the TBEV/LIV-I. ricinus PPI network, and indeed of any PPI network involving a tick-borne virus and its tick vector. While further investigation will be needed to elucidate the role of each tick protein in the replication cycle of tick-borne flaviviruses, our study provides a foundation for understanding the vector competence of I. ricinus at the molecular level. Indeed, certain PPIs may represent molecular determinants of vector competence of I. ricinus for TBEV and LIV, and potentially for other tick-borne flaviviruses.![]() Supplementary Information The online version contains supplementary material available at 10.1186/s13071-021-04651-3.
Collapse
Affiliation(s)
- Manon Lemasson
- UMR 1161 Virologie Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, Paris-Est Sup, Maisons-Alfort, France
| | - Grégory Caignard
- UMR 1161 Virologie Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, Paris-Est Sup, Maisons-Alfort, France
| | - Yves Unterfinger
- UMR 1161 Virologie Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, Paris-Est Sup, Maisons-Alfort, France
| | - Houssam Attoui
- UMR 1161 Virologie Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, Paris-Est Sup, Maisons-Alfort, France
| | - Lesley Bell-Sakyi
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Edouard Hirchaud
- Viral Genetic and Biosecurity Unit, Ploufragan-Plouzané-Niort Laboratory, ANSES, Ploufragan, France
| | - Sara Moutailler
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, Paris-Est Sup, Maisons-Alfort, France
| | | | - Damien Vitour
- UMR 1161 Virologie Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, Paris-Est Sup, Maisons-Alfort, France
| | - Jennifer Richardson
- UMR 1161 Virologie Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, Paris-Est Sup, Maisons-Alfort, France
| | - Sandrine A Lacour
- UMR 1161 Virologie Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, Paris-Est Sup, Maisons-Alfort, France.
| |
Collapse
|
23
|
Kim TK, Tirloni L, Bencosme-Cuevas E, Kim TH, Diedrich JK, Yates JR, Mulenga A. Borrelia burgdorferi infection modifies protein content in saliva of Ixodes scapularis nymphs. BMC Genomics 2021; 22:152. [PMID: 33663385 PMCID: PMC7930271 DOI: 10.1186/s12864-021-07429-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Lyme disease (LD) caused by Borrelia burgdorferi is the most prevalent tick-borne disease. There is evidence that vaccines based on tick proteins that promote tick transmission of B. burgdorferi could prevent LD. As Ixodes scapularis nymph tick bites are responsible for most LD cases, this study sought to identify nymph tick saliva proteins associated with B. burgdorferi transmission using LC-MS/MS. Tick saliva was collected using a non-invasive method of stimulating ticks (uninfected and infected: unfed, and every 12 h during feeding through 72 h, and fully-fed) to salivate into 2% pilocarpine-PBS for protein identification using LC-MS/MS. RESULTS We identified a combined 747 tick saliva proteins of uninfected and B. burgdorferi infected ticks that were classified into 25 functional categories: housekeeping-like (48%), unknown function (18%), protease inhibitors (9%), immune-related (6%), proteases (8%), extracellular matrix (7%), and small categories that account for <5% each. Notably, B. burgdorferi infected ticks secreted high number of saliva proteins (n=645) than uninfected ticks (n=376). Counter-intuitively, antimicrobial peptides, which function to block bacterial infection at tick feeding site were suppressed 23-85 folds in B. burgdorferi infected ticks. Similar to glycolysis enzymes being enhanced in mammalian cells exposed to B. burgdorferi : eight of the 10-glycolysis pathway enzymes were secreted at high abundance by B. burgdorferi infected ticks. Of significance, rabbits exposed to B. burgdorferi infected ticks acquired potent immunity that caused 40-60% mortality of B. burgdorferi infected ticks during the second infestation compared to 15-28% for the uninfected. This might be explained by ELISA data that show that high expression levels of immunogenic proteins in B. burgdorferi infected ticks. CONCLUSION Data here suggest that B. burgdorferi infection modified protein content in tick saliva to promote its survival at the tick feeding site. For instance, enzymes; copper/zinc superoxide dismutase that led to production of H2O2 that is toxic to B. burgdorferi were suppressed, while, catalase and thioredoxin that neutralize H2O2, and pyruvate kinase which yields pyruvate that protects Bb from H2O2 killing were enhanced. We conclude data here is an important resource for discovery of effective antigens for a vaccine to prevent LD.
Collapse
Affiliation(s)
- Tae Kwon Kim
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
- Department of Diagnostic Medicine and Veterinary Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, United States of America
| | - Lucas Tirloni
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
- Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, Hamilton, Montana, United States of America
| | - Emily Bencosme-Cuevas
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
| | - Tae Heung Kim
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
| | - Jolene K Diedrich
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
- Mass Spectrometry Core, Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - John R Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Albert Mulenga
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America.
| |
Collapse
|
24
|
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: 66] [Impact Index Per Article: 16.5] [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
|
25
|
Hart CE, Thangamani S. Tick-virus interactions: Current understanding and future perspectives. Parasite Immunol 2021; 43:e12815. [PMID: 33368375 DOI: 10.1111/pim.12815] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/30/2022]
Abstract
Ticks are the primary vector of arboviruses in temperate climates worldwide. They are both the vector of these pathogens to humans and an integral component of the viral sylvatic cycle. Understanding the tick-pathogen interaction provides information about the natural maintenance of these pathogens and informs the development of countermeasures against human infection. In this review, we discuss currently available information on tick-viral interactions within the broader scope of general tick immunology. While the tick immune response to several pathogens has been studied extensively, minimal work centres on responses to viral infection. This is largely due to the high pathogenicity of tick-borne viruses; this necessitates high-containment laboratories or low-pathogenicity substitute viruses. This has biased most research towards tick-borne flaviviruses. More work is required to fully understand the role of tick-virus interaction in sylvatic cycling and transmission of diverse tick-borne viruses.
Collapse
Affiliation(s)
- Charles Edward Hart
- Institute for Global Health and Translational Science, Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Saravanan Thangamani
- Institute for Global Health and Translational Science, Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
26
|
Pal U, Kitsou C, Drecktrah D, Yaş ÖB, Fikrig E. Interactions Between Ticks and Lyme Disease Spirochetes. Curr Issues Mol Biol 2020; 42:113-144. [PMID: 33289683 PMCID: PMC8045411 DOI: 10.21775/cimb.042.113] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Borrelia burgdorferi sensu lato causes Lyme borreliosis in a variety of animals and humans. These atypical bacterial pathogens are maintained in a complex enzootic life cycle that primarily involves a vertebrate host and Ixodes spp. ticks. In the Northeastern United States, I. scapularis is the main vector, while wild rodents serve as the mammalian reservoir host. As B. burgdorferi is transmitted only by I. scapularis and closely related ticks, the spirochete-tick interactions are thought to be highly specific. Various borrelial and arthropod proteins that directly or indirectly contribute to the natural cycle of B. burgdorferi infection have been identified. Discrete molecular interactions between spirochetes and tick components also have been discovered, which often play critical roles in pathogen persistence and transmission by the arthropod vector. This review will focus on the past discoveries and future challenges that are relevant to our understanding of the molecular interactions between B. burgdorferi and Ixodes ticks. This information will not only impact scientific advancements in the research of tick- transmitted infections but will also contribute to the development of novel preventive measures that interfere with the B. burgdorferi life cycle.
Collapse
Affiliation(s)
- Utpal Pal
- Department of Veterinary Medicine, University of Maryland, 8075 Greenmead Drive, College Park, MD 20742, USA
- Virginia-Maryland College of Veterinary Medicine, 8075 Greenmead Drive, College Park, MD 20742, USA
| | - Chrysoula Kitsou
- Department of Veterinary Medicine, University of Maryland, 8075 Greenmead Drive, College Park, MD 20742, USA
| | - Dan Drecktrah
- Division of Biological Sciences, University of Montana, Missoula, MT, 59812, USA
| | - Özlem Büyüktanir Yaş
- Department of Microbiology and Clinical Microbiology, Faculty of Medicine, Istinye University, Zeytinburnu, İstanbul, 34010, Turkey
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
27
|
Abstract
Increases in tick-borne disease prevalence and transmission are important public health issues. Efforts to control these emerging diseases are frustrated by the struggle to control tick populations and to detect and treat infections caused by the pathogens that they transmit. This review covers tick-borne infectious diseases of nonrickettsial bacterial, parasitic, and viral origins. While tick surveillance and tracking inform our understanding of the importance of the spread and ecology of ticks and help identify areas of risk for disease transmission, the vectors are not the focus of this document. Here, we emphasize the most significant pathogens that infect humans as well as the epidemiology, clinical features, diagnosis, and treatment of diseases that they cause. Although detection via molecular or immunological methods has improved, tick-borne diseases continue to remain underdiagnosed, making the scope of the problem difficult to assess. Our current understanding of the incidence of tick-borne diseases is discussed in this review. An awareness of the diseases that can be transmitted by ticks in specific locations is key to detection and selection of appropriate treatment. As tick-transmitted pathogens are discovered and emerge in new geographic regions, our ability to detect, describe, and understand the growing public health threat must also grow to meet the challenge.
Collapse
|
28
|
Hernandez EP, Talactac MR, Fujisaki K, Tanaka T. The case for oxidative stress molecule involvement in the tick-pathogen interactions -an omics approach. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 100:103409. [PMID: 31200008 DOI: 10.1016/j.dci.2019.103409] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 06/09/2023]
Abstract
The blood-feeding behavior of ticks has resulted in them becoming one of the most important vectors of disease-causing pathogens. Ticks possess a well-developed innate immune system to counter invading pathogens. However, the coevolution of ticks with tick-borne pathogens has adapted these pathogens to the tick's physiology and immune response through several mechanisms including transcriptional regulation. The recent development in tick and tick-borne disease research greatly involved the "omics" approach. The omics approach takes a look en masse at the different genes, proteins, metabolomes, and the microbiome of the ticks that could be differentiated during pathogen infection. Data from this approach revealed that oxidative stress-related molecules in ticks are differentiated and possibly being exploited by the pathogens to evade the tick's immune response. In this study, we review and discuss transcriptomic and proteomic data for some oxidative stress molecules differentially expressed during pathogen infection. We also discuss metabolomics and microbiome data as well as functional genomics in order to provide insight into the tick-pathogen interaction.
Collapse
Affiliation(s)
- Emmanuel Pacia Hernandez
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0056, Japan; Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi, 753-8515, Japan
| | - Melbourne Rio Talactac
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0056, Japan; Department of Clinical and Population Health, College of Veterinary Medicine and Biomedical Sciences, Cavite State University, Cavite, 4122, Philippines
| | - Kozo Fujisaki
- National Agricultural and Food Research Organization, 3-1-5 Kannondai, Tsukuba, Ibaraki, 305-0856, Japan
| | - Tetsuya Tanaka
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0056, Japan; Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi, 753-8515, Japan.
| |
Collapse
|
29
|
Loginov DS, Loginova YF, Dycka F, Böttinger K, Vechtova P, Sterba J. Tissue-specific signatures in tick cell line MS profiles. Parasit Vectors 2019; 12:212. [PMID: 31060584 PMCID: PMC6503378 DOI: 10.1186/s13071-019-3460-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/27/2019] [Indexed: 11/10/2022] Open
Abstract
Background The availability of tick in vitro cell culture systems has facilitated many aspects of tick research, including proteomics. However, certain cell lines have shown a tissue-specific response to infection. Thus, a more thorough characterization of tick cell lines is necessary. Proteomic comparative studies of various tick cell lines will contribute to more efficient application of tick cell lines as model systems for investigation of host-vector-pathogen interactions. Results Three cell lines obtained from a hard tick, Ixodes ricinus, and two from I. scapularis were investigated. A cell mass spectrometry approach (MALDI-TOF MS) was applied, as well as classical proteomic workflows. Using PCA, tick cell line MS profiles were grouped into three clusters comprising IRE/CTVM19 and ISE18, IRE11 and IRE/CTVM20, and ISE6 cell lines. Two other approaches confirmed the results of PCA: in-solution digestion followed by nanoLC-ESI-Q-TOF MS/MS and 2D electrophoresis. The comparison of MS spectra of the cell lines and I. ricinus tick organs revealed 29 shared peaks. Of these, five were specific for ovaries, three each for gut and salivary glands, and one for Malpighian tubules. For the first time, characteristic peaks in MS profiles of tick cell lines were assigned to proteins identified in acidic extracts of corresponding cell lines. Conclusions Several organ-specific MS signals were revealed in the profiles of tick cell lines. Electronic supplementary material The online version of this article (10.1186/s13071-019-3460-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dmitry S Loginov
- Faculty of Science, University of South Bohemia, Branišovská 1760, 37005, Ceske Budejovice, Czech Republic. .,Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 1760, 37005, Ceske Budejovice, Czech Republic.
| | - Yana F Loginova
- Faculty of Science, University of South Bohemia, Branišovská 1760, 37005, Ceske Budejovice, Czech Republic.,Orekhovich Institute of Biomedical Chemistry, Pogodinskaja str. 10, Moscow, 119191, Russia
| | - Filip Dycka
- Faculty of Science, University of South Bohemia, Branišovská 1760, 37005, Ceske Budejovice, Czech Republic
| | - Katharina Böttinger
- Faculty of Science, University of South Bohemia, Branišovská 1760, 37005, Ceske Budejovice, Czech Republic
| | - Pavlina Vechtova
- Faculty of Science, University of South Bohemia, Branišovská 1760, 37005, Ceske Budejovice, Czech Republic.,Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 1760, 37005, Ceske Budejovice, Czech Republic
| | - Jan Sterba
- Faculty of Science, University of South Bohemia, Branišovská 1760, 37005, Ceske Budejovice, Czech Republic.,Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branišovská 1760, 37005, Ceske Budejovice, Czech Republic
| |
Collapse
|
30
|
Barros-Battesti DM, Machado RZ, André MR, de Sousa KCM, Franze DA, Lima-Duarte L, Cirelli-Moraes A, Nunes PH, Labruna MB, Moraes-Filho J, Martins MM, Szabó MPJ. Successful Infection of Tick Cell Cultures of Rhipicephalus sanguineus (Tropical Lineage) with Ehrlichia canis. Vector Borne Zoonotic Dis 2018; 18:653-662. [PMID: 30222504 DOI: 10.1089/vbz.2017.2197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
There are two distinct lineages of ticks, Rhipicephalus sanguineus, in South America: tropical and temperate lineages. Only the tropical lineage is recognized as competent vector for Ehrlichia canis. The epidemiological data of canine monocytic ehrlichiosis is congruent with the distribution of the two lineages of R. sanguineus. Herein, we report the infection of R. sanguineus (tropical lineage) cell cultures with E. canis, after cryopreservation. R. sanguineus (tropical lineage) cell identity was confirmed by sequencing using a 16S rDNA gene fragment. Tick cell cultures were prepared in L-15B medium supplemented with 10%, 15%, and 20% Fetal Bovine Serum (FBS), and 10% of Tryptose Phosphate Broth (TPB). Cell cultures developed better at the concentration of 20% of FBS. Cultures in the fifth harvest (approximately 7 months later) were selected for the first infections. Optimal R. sanguineus cell growth and adhesion was observed (5.0 × 106 cells/mL, and the population doubling time every 57 h). Once infected with E. canis, the cultures were maintained in L-15B medium supplemented with 2% and 5% of FBS fortified with iron and 10% TPB. Infected cells were also cryopreserved. DNA was extracted from infected and noninfected cells and analyzed using quantitative real-time PCR targeting the E. canis-dsb gene. Primary culture of the fifth passage was infected by E. canis and it maintained the pathogen for at least 40 days before partial cell destruction. Subcultures of infected cells (fresh and cryopreserved cultures) onto new tick cell cultures were successful. The E. canis infection was confirmed by real-time PCR and light and transmission electron microscopy. The R. sanguineus (tropical lineage) cells infected with E. canis successfully infected new tick cell cultures, showing that these cells could be an alternative substrate for maintenance of this pathogen.
Collapse
Affiliation(s)
- Darci Moraes Barros-Battesti
- 1 Department of Veterinary Pathology, Faculty of Agricultural and Veterinary Sciences, State University Julio de Mesquita Filho (UNESP) , Jaboticabal, Brazil
- 2 Laboratory of Parasitology, Butantan Institute , São Paulo, Brazil
| | - Rosangela Zacarias Machado
- 1 Department of Veterinary Pathology, Faculty of Agricultural and Veterinary Sciences, State University Julio de Mesquita Filho (UNESP) , Jaboticabal, Brazil
| | - Marcos Rogério André
- 1 Department of Veterinary Pathology, Faculty of Agricultural and Veterinary Sciences, State University Julio de Mesquita Filho (UNESP) , Jaboticabal, Brazil
| | - Keyla Carstens Marques de Sousa
- 1 Department of Veterinary Pathology, Faculty of Agricultural and Veterinary Sciences, State University Julio de Mesquita Filho (UNESP) , Jaboticabal, Brazil
| | | | | | | | - Pablo Henrique Nunes
- 3 Federal University of Latin American Integration-UNILA , Foz do Iguaçu, Brazil
| | - Marcelo Bahia Labruna
- 4 Department of Preventive Veterinary Medicine and Animal Science, School of Veterinary Medicine, University of São Paulo , São Paulo, Brazil
| | - Jonas Moraes-Filho
- 4 Department of Preventive Veterinary Medicine and Animal Science, School of Veterinary Medicine, University of São Paulo , São Paulo, Brazil
- 5 Veterinary Medicine, University of Santo Amaro , São Paulo, Brazil
| | | | | |
Collapse
|
31
|
Khanal S, Taank V, Anderson JF, Sultana H, Neelakanta G. Arthropod transcriptional activator protein-1 (AP-1) aids tick-rickettsial pathogen survival in the cold. Sci Rep 2018; 8:11409. [PMID: 30061607 PMCID: PMC6065373 DOI: 10.1038/s41598-018-29654-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/12/2018] [Indexed: 11/09/2022] Open
Abstract
Ixodes scapularis ticks transmit several pathogens to humans including rickettsial bacterium, Anaplasma phagocytophilum. Here, we report that A. phagocytophilum uses tick transcriptional activator protein-1 (AP-1) as a molecular switch in the regulation of arthropod antifreeze gene, iafgp. RNAi-mediated silencing of ap-1 expression significantly affected iafgp gene expression and A. phagocytophilum burden in ticks upon acquisition from the murine host. Gel shift assays provide evidence that both the bacterium and AP-1 influences iafgp promoter and expression. The luciferase assays revealed that a region of approximately 700 bp upstream of the antifreeze gene is sufficient for AP-1 binding to promote iafgp gene expression. Furthermore, survival assays revealed that AP-1-deficient ticks were more susceptible to cold in comparison to the mock controls. In addition, this study also indicates arthropod AP-1 as a global regulator for some of the tick genes critical for A. phagocytophilum survival in the vector. In summary, our study defines a novel mode of arthropod signaling for the survival of both rickettsial pathogen and its medically important vector in the cold.
Collapse
Affiliation(s)
- Supreet Khanal
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - Vikas Taank
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - John F Anderson
- Department of Entomology, Connecticut Agricultural Experiment Station, New Haven, CT, USA
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
- Center for Molecular Medicine, Old Dominion University, Norfolk, VA, USA
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA.
- Center for Molecular Medicine, Old Dominion University, Norfolk, VA, USA.
| |
Collapse
|
32
|
De La Fuente J, Villar M, Estrada-Peña A, Olivas JA. High throughput discovery and characterization of tick and pathogen vaccine protective antigens using vaccinomics with intelligent Big Data analytic techniques. Expert Rev Vaccines 2018; 17:569-576. [DOI: 10.1080/14760584.2018.1493928] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- José De La Fuente
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, USA
| | - Margarita Villar
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ciudad Real, Spain
| | | | - José A. Olivas
- Technologies and Information Systems Institute UCLM, Ciudad Real, Spain
| |
Collapse
|
33
|
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
|
34
|
Espinosa PJ, Alberdi P, Villar M, Cabezas-Cruz A, de la Fuente J. Heat Shock Proteins in Vector-pathogen Interactions: The Anaplasma phagocytophilum Model. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/978-3-319-73377-7_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
Grabowski JM, Hill CA. A Roadmap for Tick-Borne Flavivirus Research in the "Omics" Era. Front Cell Infect Microbiol 2017; 7:519. [PMID: 29312896 PMCID: PMC5744076 DOI: 10.3389/fcimb.2017.00519] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/05/2017] [Indexed: 12/30/2022] Open
Abstract
Tick-borne flaviviruses (TBFs) affect human health globally. Human vaccines provide protection against some TBFs, and antivirals are available, yet TBF-specific control strategies are limited. Advances in genomics offer hope to understand the viral complement transmitted by ticks, and to develop disruptive, data-driven technologies for virus detection, treatment, and control. The genome assemblies of Ixodes scapularis, the North American tick vector of the TBF, Powassan virus, and other tick vectors, are providing insights into tick biology and pathogen transmission and serve as nucleation points for expanded genomic research. Systems biology has yielded insights to the response of tick cells to viral infection at the transcript and protein level, and new protein targets for vaccines to limit virus transmission. Reverse vaccinology approaches have moved candidate tick antigenic epitopes into vaccine development pipelines. Traditional drug and in silico screening have identified candidate antivirals, and target-based approaches have been developed to identify novel acaricides. Yet, additional genomic resources are required to expand TBF research. Priorities include genome assemblies for tick vectors, “omic” studies involving high consequence pathogens and vectors, and emphasizing viral metagenomics, tick-virus metabolomics, and structural genomics of TBF and tick proteins. Also required are resources for forward genetics, including the development of tick strains with quantifiable traits, genetic markers and linkage maps. Here we review the current state of genomic research on ticks and tick-borne viruses with an emphasis on TBFs. We outline an ambitious 10-year roadmap for research in the “omics era,” and explore key milestones needed to accomplish the goal of delivering three new vaccines, antivirals and acaricides for TBF control by 2030.
Collapse
Affiliation(s)
- Jeffrey M Grabowski
- Biology of Vector-Borne Viruses Section, Laboratory of Virology, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Catherine A Hill
- Department of Entomology, Purdue University, West Lafayette, IN, United States.,Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
36
|
Kazimírová M, Thangamani S, Bartíková P, Hermance M, Holíková V, Štibrániová I, Nuttall PA. Tick-Borne Viruses and Biological Processes at the Tick-Host-Virus Interface. Front Cell Infect Microbiol 2017; 7:339. [PMID: 28798904 PMCID: PMC5526847 DOI: 10.3389/fcimb.2017.00339] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/11/2017] [Indexed: 01/08/2023] Open
Abstract
Ticks are efficient vectors of arboviruses, although less than 10% of tick species are known to be virus vectors. Most tick-borne viruses (TBV) are RNA viruses some of which cause serious diseases in humans and animals world-wide. Several TBV impacting human or domesticated animal health have been found to emerge or re-emerge recently. In order to survive in nature, TBV must infect and replicate in both vertebrate and tick cells, representing very different physiological environments. Information on molecular mechanisms that allow TBV to switch between infecting and replicating in tick and vertebrate cells is scarce. In general, ticks succeed in completing their blood meal thanks to a plethora of biologically active molecules in their saliva that counteract and modulate different arms of the host defense responses (haemostasis, inflammation, innate and acquired immunity, and wound healing). The transmission of TBV occurs primarily during tick feeding and is a complex process, known to be promoted by tick saliva constituents. However, the underlying molecular mechanisms of TBV transmission are poorly understood. Immunomodulatory properties of tick saliva helping overcome the first line of defense to injury and early interactions at the tick-host skin interface appear to be essential in successful TBV transmission and infection of susceptible vertebrate hosts. The local host skin site of tick attachment, modulated by tick saliva, is an important focus of virus replication. Immunomodulation of the tick attachment site also promotes co-feeding transmission of viruses from infected to non-infected ticks in the absence of host viraemia (non-viraemic transmission). Future research should be aimed at identification of the key tick salivary molecules promoting virus transmission, and a molecular description of tick-host-virus interactions and of tick-mediated skin immunomodulation. Such insights will enable the rationale design of anti-tick vaccines that protect against disease caused by tick-borne viruses.
Collapse
Affiliation(s)
- Mária Kazimírová
- Department of Medical Zoology, Institute of Zoology, Slovak Academy of SciencesBratislava, Slovakia
| | - Saravanan Thangamani
- Department of Pathology, University of Texas Medical BranchGalveston, TX, United States
- Institute for Human Infections and Immunity, University of Texas Medical BranchGalveston, TX, United States
- Center for Tropical Diseases, University of Texas Medical BranchGalveston, TX, United States
| | - Pavlína Bartíková
- Biomedical Research Center, Institute of Virology, Slovak Academy of SciencesBratislava, Slovakia
| | - Meghan Hermance
- Department of Pathology, University of Texas Medical BranchGalveston, TX, United States
- Institute for Human Infections and Immunity, University of Texas Medical BranchGalveston, TX, United States
- Center for Tropical Diseases, University of Texas Medical BranchGalveston, TX, United States
| | - Viera Holíková
- Biomedical Research Center, Institute of Virology, Slovak Academy of SciencesBratislava, Slovakia
| | - Iveta Štibrániová
- Biomedical Research Center, Institute of Virology, Slovak Academy of SciencesBratislava, Slovakia
| | - Patricia A. Nuttall
- Department of Zoology, University of OxfordOxford, United Kingdom
- Centre for Ecology and HydrologyWallingford, United Kingdom
| |
Collapse
|
37
|
Mansfield KL, Jizhou L, Phipps LP, Johnson N. Emerging Tick-Borne Viruses in the Twenty-First Century. Front Cell Infect Microbiol 2017; 7:298. [PMID: 28744449 PMCID: PMC5504652 DOI: 10.3389/fcimb.2017.00298] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/19/2017] [Indexed: 12/18/2022] Open
Abstract
Ticks, as a group, are second only to mosquitoes as vectors of pathogens to humans and are the primary vector for pathogens of livestock, companion animals, and wildlife. The role of ticks in the transmission of viruses has been known for over 100 years and yet new pathogenic viruses are still being detected and known viruses are continually spreading to new geographic locations. Partly as a result of their novelty, tick-virus interactions are at an early stage in understanding. For some viruses, even the principal tick-vector is not known. It is likely that tick-borne viruses will continue to emerge and challenge public and veterinary health long into the twenty-first century. However, studies focusing on tick saliva, a critical component of tick feeding, virus transmission, and a target for control of ticks and tick-borne diseases, point toward solutions to emerging viruses. The aim of this review is to describe some currently emerging tick-borne diseases, their causative viruses, and to discuss research on virus-tick interactions. Through focus on this area, future protein targets for intervention and vaccine development may be identified.
Collapse
Affiliation(s)
- Karen L Mansfield
- Animal and Plant Health AgencyAddlestone, United Kingdom.,Institute of Infection and Global Health, University of LiverpoolLiverpool, United Kingdom
| | - Lv Jizhou
- Animal and Plant Health AgencyAddlestone, United Kingdom.,Chinese Academy of Inspection and QuarantineBeijing, China
| | - L Paul Phipps
- Animal and Plant Health AgencyAddlestone, United Kingdom
| | - Nicholas Johnson
- Animal and Plant Health AgencyAddlestone, United Kingdom.,Faculty of Health and Medicine, University of SurreyGuildford, United Kingdom
| |
Collapse
|
38
|
Contreras M, Alberdi P, Mateos-Hernández L, Fernández de Mera IG, García-Pérez AL, Vancová M, Villar M, Ayllón N, Cabezas-Cruz A, Valdés JJ, Stuen S, Gortazar C, de la Fuente J. Anaplasma phagocytophilum MSP4 and HSP70 Proteins Are Involved in Interactions with Host Cells during Pathogen Infection. Front Cell Infect Microbiol 2017; 7:307. [PMID: 28725639 PMCID: PMC5496961 DOI: 10.3389/fcimb.2017.00307] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 06/20/2017] [Indexed: 12/18/2022] Open
Abstract
Anaplasma phagocytophilum transmembrane and surface proteins play a role during infection and multiplication in host neutrophils and tick vector cells. Recently, A. phagocytophilum Major surface protein 4 (MSP4) and Heat shock protein 70 (HSP70) were shown to be localized on the bacterial membrane, with a possible role during pathogen infection in ticks. In this study, we hypothesized that A. phagocytophilum MSP4 and HSP70 have similar functions in tick-pathogen and host-pathogen interactions. To address this hypothesis, herein we characterized the role of these bacterial proteins in interaction and infection of vertebrate host cells. The results showed that A. phagocytophilum MSP4 and HSP70 are involved in host-pathogen interactions, with a role for HSP70 during pathogen infection. The analysis of the potential protective capacity of MSP4 and MSP4-HSP70 antigens in immunized sheep showed that MSP4-HSP70 was only partially protective against pathogen infection. This limited protection may be associated with several factors, including the recognition of non-protective epitopes by IgG in immunized lambs. Nevertheless, these antigens may be combined with other candidate protective antigens for the development of vaccines for the control of human and animal granulocytic anaplasmosis. Focusing on the characterization of host protective immune mechanisms and protein-protein interactions at the host-pathogen interface may lead to the discovery and design of new effective protective antigens.
Collapse
Affiliation(s)
- Marinela Contreras
- SaBio, Instituto de Investigación en Recursos Cinegéticos, Consejo Superior de Investigaciones Científicas, CSIC-UCLM-JCCMCiudad Real, Spain
| | - Pilar Alberdi
- SaBio, Instituto de Investigación en Recursos Cinegéticos, Consejo Superior de Investigaciones Científicas, CSIC-UCLM-JCCMCiudad Real, Spain
| | - Lourdes Mateos-Hernández
- SaBio, Instituto de Investigación en Recursos Cinegéticos, Consejo Superior de Investigaciones Científicas, CSIC-UCLM-JCCMCiudad Real, Spain
| | - Isabel G Fernández de Mera
- SaBio, Instituto de Investigación en Recursos Cinegéticos, Consejo Superior de Investigaciones Científicas, CSIC-UCLM-JCCMCiudad Real, Spain
| | - Ana L García-Pérez
- Departamento de Sanidad Animal, Instituto Vasco de Investigación y Desarrollo Agrario (NEIKER)Derio, Spain
| | - Marie Vancová
- Biology Centre, Czech Academy of Sciences, Institute of ParasitologyČeské Budějovice, Czechia.,Faculty of Science, University of South BohemiaČeské Budějovice, Czechia
| | - Margarita Villar
- SaBio, Instituto de Investigación en Recursos Cinegéticos, Consejo Superior de Investigaciones Científicas, CSIC-UCLM-JCCMCiudad Real, Spain
| | - Nieves Ayllón
- SaBio, Instituto de Investigación en Recursos Cinegéticos, Consejo Superior de Investigaciones Científicas, CSIC-UCLM-JCCMCiudad Real, Spain
| | - Alejandro Cabezas-Cruz
- Biology Centre, Czech Academy of Sciences, Institute of ParasitologyČeské Budějovice, Czechia.,Faculty of Science, University of South BohemiaČeské Budějovice, Czechia.,UMR BIPAR, Animal Health Laboratory, INRA, ANSES, ENVAMaisons Alfort, France
| | - James J Valdés
- Biology Centre, Czech Academy of Sciences, Institute of ParasitologyČeské Budějovice, Czechia.,Department of Virology, Veterinary Research InstituteBrno, Czechia
| | - Snorre Stuen
- Department of Production Animal Clinical Sciences, Norwegian University of Life SciencesSandnes, Norway
| | - Christian Gortazar
- SaBio, Instituto de Investigación en Recursos Cinegéticos, Consejo Superior de Investigaciones Científicas, CSIC-UCLM-JCCMCiudad Real, Spain
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos, Consejo Superior de Investigaciones Científicas, CSIC-UCLM-JCCMCiudad Real, Spain.,Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State UniversityStillwater, OK, United States
| |
Collapse
|
39
|
Abstract
Ticks are important vectors of viruses that infect and cause disease in man, livestock, and companion animals. The major focus of investigation of tick-borne viruses has been the interaction with the mammalian host, particularly the mechanisms underlying disease and the development of vaccines to prevent infection. Only recently has research begun to investigate the interaction of the virus with the tick host. This is striking when considering that the virus spends far more time infecting the tick vector relative to the vertebrate host. The assumption has been that the tick host and virus have evolved to reach an equilibrium whereby virus infection does not impede the tick life cycle and conversely, the tick does not restrict virus replication and through blood-feeding on vertebrates, disseminates the virus. The development and application of new technologies to tick-pathogen interactions has been fuelled by a number of developments in recent years. This includes the release of the first draft of a tick genome, that of Ixodes scapularis, and the availability of tick-cell lines as convenient models to investigate interactions. One of the by-products of these investigations has been the observation of familiar proteins in new situations. One such protein family is Toll and Toll-like receptors that in vertebrates play a key role in detection of microorganisms, including viruses. But does Toll signaling play a similar role in detection of virus infection in ticks, and if it does, how does this affect the maintenance of viruses within the tick?
Collapse
Affiliation(s)
- Nicholas Johnson
- Animal and Plant Health AgencyAddlestone, United Kingdom
- Faculty of Health and Medicine, University of SurreyGuildford, United Kingdom
| |
Collapse
|
40
|
de la Fuente J, Antunes S, Bonnet S, Cabezas-Cruz A, Domingos AG, Estrada-Peña A, Johnson N, Kocan KM, Mansfield KL, Nijhof AM, Papa A, Rudenko N, Villar M, Alberdi P, Torina A, Ayllón N, Vancova M, Golovchenko M, Grubhoffer L, Caracappa S, Fooks AR, Gortazar C, Rego ROM. Tick-Pathogen Interactions and Vector Competence: Identification of Molecular Drivers for Tick-Borne Diseases. Front Cell Infect Microbiol 2017; 7:114. [PMID: 28439499 PMCID: PMC5383669 DOI: 10.3389/fcimb.2017.00114] [Citation(s) in RCA: 271] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/22/2017] [Indexed: 01/10/2023] Open
Abstract
Ticks and the pathogens they transmit constitute a growing burden for human and animal health worldwide. Vector competence is a component of vectorial capacity and depends on genetic determinants affecting the ability of a vector to transmit a pathogen. These determinants affect traits such as tick-host-pathogen and susceptibility to pathogen infection. Therefore, the elucidation of the mechanisms involved in tick-pathogen interactions that affect vector competence is essential for the identification of molecular drivers for tick-borne diseases. In this review, we provide a comprehensive overview of tick-pathogen molecular interactions for bacteria, viruses, and protozoa affecting human and animal health. Additionally, the impact of tick microbiome on these interactions was considered. Results show that different pathogens evolved similar strategies such as manipulation of the immune response to infect vectors and facilitate multiplication and transmission. Furthermore, some of these strategies may be used by pathogens to infect both tick and mammalian hosts. Identification of interactions that promote tick survival, spread, and pathogen transmission provides the opportunity to disrupt these interactions and lead to a reduction in tick burden and the prevalence of tick-borne diseases. Targeting some of the similar mechanisms used by the pathogens for infection and transmission by ticks may assist in development of preventative strategies against multiple tick-borne diseases.
Collapse
Affiliation(s)
- José de la Fuente
- SaBio. Instituto de Investigación en Recursos Cinegéticos CSIC-UCLM-JCCMCiudad Real, Spain.,Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State UniversityStillwater, OK, USA
| | - Sandra Antunes
- Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de LisboaLisboa, Portugal
| | | | - Alejandro Cabezas-Cruz
- UMR BIPAR INRA-ANSES-ENVAMaisons-Alfort, France.,Biology Centre, Czech Academy of Sciences, Institute of ParasitologyCeske Budejovice, Czechia.,Faculty of Science, University of South BohemiaČeské Budějovice, Czechia
| | - Ana G Domingos
- Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de LisboaLisboa, Portugal
| | | | - Nicholas Johnson
- Animal and Plant Health AgencySurrey, UK.,Faculty of Health and Medicine, University of SurreyGuildford, UK
| | - Katherine M Kocan
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State UniversityStillwater, OK, USA
| | - Karen L Mansfield
- Animal and Plant Health AgencySurrey, UK.,Institute of Infection and Global Health, University of LiverpoolLiverpool, UK
| | - Ard M Nijhof
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität BerlinBerlin, Germany
| | - Anna Papa
- Department of Microbiology, Medical School, Aristotle University of ThessalonikiThessaloniki, Greece
| | - Nataliia Rudenko
- Biology Centre, Czech Academy of Sciences, Institute of ParasitologyCeske Budejovice, Czechia
| | - Margarita Villar
- SaBio. Instituto de Investigación en Recursos Cinegéticos CSIC-UCLM-JCCMCiudad Real, Spain
| | - Pilar Alberdi
- SaBio. Instituto de Investigación en Recursos Cinegéticos CSIC-UCLM-JCCMCiudad Real, Spain
| | - Alessandra Torina
- National Center of Reference for Anaplasma, Babesia, Rickettsia and Theileria, Intituto Zooprofilattico Sperimentale della SiciliaSicily, Italy
| | - Nieves Ayllón
- SaBio. Instituto de Investigación en Recursos Cinegéticos CSIC-UCLM-JCCMCiudad Real, Spain
| | - Marie Vancova
- Biology Centre, Czech Academy of Sciences, Institute of ParasitologyCeske Budejovice, Czechia
| | - Maryna Golovchenko
- Biology Centre, Czech Academy of Sciences, Institute of ParasitologyCeske Budejovice, Czechia
| | - Libor Grubhoffer
- Biology Centre, Czech Academy of Sciences, Institute of ParasitologyCeske Budejovice, Czechia.,Faculty of Science, University of South BohemiaČeské Budějovice, Czechia
| | - Santo Caracappa
- National Center of Reference for Anaplasma, Babesia, Rickettsia and Theileria, Intituto Zooprofilattico Sperimentale della SiciliaSicily, Italy
| | - Anthony R Fooks
- Animal and Plant Health AgencySurrey, UK.,Institute of Infection and Global Health, University of LiverpoolLiverpool, UK
| | - Christian Gortazar
- SaBio. Instituto de Investigación en Recursos Cinegéticos CSIC-UCLM-JCCMCiudad Real, Spain
| | - Ryan O M Rego
- Biology Centre, Czech Academy of Sciences, Institute of ParasitologyCeske Budejovice, Czechia.,Faculty of Science, University of South BohemiaČeské Budějovice, Czechia
| |
Collapse
|