1
|
Turck JW, Sultana H, Neelakanta G. Arthropod autophagy molecules facilitate Anaplasma phagocytophilum infection of Ixodes scapularis tick cells. Commun Biol 2025; 8:433. [PMID: 40082564 PMCID: PMC11906822 DOI: 10.1038/s42003-025-07859-6] [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: 08/16/2024] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
Ixodes scapularis ticks transmit several medically important pathogens including Anaplasma phagocytophilum to humans and animals. In this study, we provide evidence that A. phagocytophilum modulates autophagy molecules for its survival in tick cells. qRT-PCR analysis revealed that A. phagocytophilum infection results in the upregulation of tyrosine phosphatase, shp-2, and serine/threonine-protein kinase, mTOR, in ticks and tick cells. RNAi-mediated knockdown of shp-2 or functional blocking with SHP-2 inhibitor resulted in significantly increased bacterial burden and reduced phospho-mTOR levels in A. phagocytophilum-infected tick cells. In addition, treatment of A. phagocytophilum-infected tick cells with rapamycin (mTOR inhibitor) resulted in significantly increased bacterial burden and reduced phospho-mTOR levels. Furthermore, expression of autophagy molecules such as atg14 and ulk1 were noted to be upregulated in both A. phagocytophilum-infected unfed ticks and tick cells. RNAi-mediated silencing of atg14 or ulk1 affected bacterial growth in tick cells. Collectively, these results not only indicate distinct host and pathogen responses in tick-A. phagocytophilum interactions but also suggest that this bacterium modulates autophagy molecules for its survival in ticks.
Collapse
Affiliation(s)
- Jeremy W Turck
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| | - Hameeda Sultana
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| | - Girish Neelakanta
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA.
| |
Collapse
|
2
|
Namjoshi P, Kolape J, Patel A, Sultana H, Neelakanta G. Rickettsial pathogen augments tick vesicular-associated membrane proteins for infection and survival in the vector host. mBio 2025; 16:e0354924. [PMID: 39950824 PMCID: PMC11898744 DOI: 10.1128/mbio.03549-24] [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: 11/18/2024] [Accepted: 01/21/2025] [Indexed: 03/14/2025] Open
Abstract
Anaplasma phagocytophilum is an obligate intracellular rickettsial pathogen that infects humans and animals. The black-legged tick Ixodes scapularis acts as a vector and transmits this bacterium to the vertebrate host. Upon entry into a host cell, A. phagocytophilum resides and multiplies in a host-derived vacuole called morulae. There is not much information available on the molecules that play an important role(s) in A. phagocytophilum entry and formation of these morulae in tick cells. In this study, we provide evidence that tick vesicular-associated membrane proteins, VAMP3 and VAMP4, play important roles in this phenomenon. Quantitative real-time polymerase chain reaction (QRT-PCR) analysis showed that both vamp3 and vamp4 transcripts are significantly upregulated at early time points of A. phagocytophilum infection in tick cells. We noted that both VAMP3 and VAMP4 predominantly localized to the A. phagocytophilum-containing vacuole. RNAi-mediated silencing of vamp3 and/or vamp4 expression, followed by confocal microscopy and expression analysis, indicated an impairment in A. phagocytophilum morulae formation in tick cells. We also noted that VAMP3 and VAMP4 play a role in the A. phagocytophilum persistent infection of ticks and tick cells. Furthermore, RNAi-mediated silencing of expression of arthropod vamp3 and vamp4 affected bacterial acquisition from an infected murine host to ticks. Collectively, this study not only provides evidence on the role of arthropod vesicular-associated membrane proteins in A. phagocytophilum morulae formation in tick cells but also demonstrates that these proteins are important for bacterial acquisition from an infected vertebrate host into ticks. IMPORTANCE Anaplasma phagocytophilum is a tick-borne pathogen primarily transmitted by black-legged Ixodes scapularis ticks to humans and animals. This bacterium enters host cells, forms a host-derived vacuole, and multiplies within this vacuole. The molecules that are critical in the formation of host-derived vacuole in tick cells is currently not well-characterized. In this study, we provide evidence that arthropod vesicular-associated membrane proteins, VAMP3 and VAMP4, are critical for A. phagocytophilum early and persistent infection in tick cells. These arthropod proteins are important for the formation of host-derived vacuoles in tick cells. Our study also provides evidence that these proteins are important for A. phagocytophilum acquisition from the infected murine host into ticks. Characterization of tick molecules important in bacterial entry and/or survival in the vector host could lead to the development of strategies to target this and perhaps other rickettsial pathogens.
Collapse
Affiliation(s)
- Prachi Namjoshi
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Jaydeep Kolape
- Advanced Microscopy and Imaging Center, College of Arts and Sciences, University of Tennessee, Knoxville, Tennessee, USA
| | - Avni Patel
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Hameeda Sultana
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Girish Neelakanta
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
3
|
Vimonish R, Capelli-Peixoto J, Johnson W, Kappmeyer L, Saelao P, Taus N, Chung C, Ueti M. Transcriptomic analysis of Rhipicephalus microplus hemocytes from female ticks infected with Babesia bovis or Babesia bigemina. Parasit Vectors 2025; 18:37. [PMID: 39901199 PMCID: PMC11789329 DOI: 10.1186/s13071-025-06662-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/07/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Tick hemolymph is a sterile fluid that carries nutrients to maintain tick health. The hemolymph creates a hostile environment for invaders including the destruction of microorganisms by its circulating hemocytes. However, Babesia parasites escape and disseminate to other organs through the hemolymph to continue their transmission life cycle. Still, it is unknown how tick hemocytes respond to B. bovis or B. bigemina infection. In this study, we conducted a transcriptomic analysis of hemocytes from female Rhipicephalus microplus ticks infected with Babesia parasites to understand how gene expression changes during parasite infection. METHODS During Babesia acute infection, female R. microplus ticks were fed on bovines to acquire parasites. Engorged females were collected and incubated to develop Babesia kinetes in tick hemolymph. The hemolymph was examined to identify ticks that were highly infected with Babesia kinetes. Hemocyte cells were collected from replete female ticks infected with Babesia bovis or Babesia bigemina to perform high-throughput RNA-sequencing (RNA-Seq) analysis. RESULTS This study identified major changes in the gene profile of tick hemocytes during Babesia infection. The main groups of hemocyte genes that were altered during Babesia infection were associated with metabolism, immunity, and cytoskeletal rearrangement. Upregulated genes were mainly involved in defense mechanisms, while downregulated genes were related to cell proliferation and apoptosis. However, the expression of hemocyte genes varied among Babesia species' infections, and it reflected the changes that occurred in the tick's physiology, including growth, reproduction, and skeletal muscle development. CONCLUSIONS The differential gene expression of R. microplus hemocytes revealed that genes highly regulated upon Babesia infection were related to metabolism, tick immunity, cell growth, apoptosis, development, metabolism, and reproduction. Additional research is necessary to further define the genes that exhibited varying expression levels in hemocytes during the infection. The findings of this study will enhance our understanding on how Babesia parasites survive in the hostile environment of ticks and perpetuate their transmission cycle, ultimately contributing to the spread of bovine babesiosis.
Collapse
Affiliation(s)
- Rubikah Vimonish
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.
| | - Janaina Capelli-Peixoto
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Wendell Johnson
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | | | - Perot Saelao
- Veterinary Pest Genetic Research Unit, USDA-ARS, Kerrville, TX, USA
| | - Naomi Taus
- Animal Disease Research Unit, USDA-ARS, Pullman, WA, USA
| | - Chungwon Chung
- Animal Disease Research Unit, USDA-ARS, Pullman, WA, USA
| | - Massaro Ueti
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
- Animal Disease Research Unit, USDA-ARS, Pullman, WA, USA
| |
Collapse
|
4
|
Zając V, Bell-Sakyi L, Wójcik-Fatla A. Use of Tick Cell Lines in Co-Infection Studies with a Preliminary Study of Co-Culture of Borrelia burgdorferi and Anaplasma phagocytophilum. Pathogens 2025; 14:78. [PMID: 39861039 PMCID: PMC11769331 DOI: 10.3390/pathogens14010078] [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: 12/06/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Ixodes ricinus is an important vector of infectious human and livestock diseases in Europe. Co-infections of pathogens in ticks and hosts have been reported. Tick cell lines offer a useful model system for study of co-infections. We present a review of the existing literature on co-infections in tick cell lines. Previous studies have demonstrated the usefulness of tick cell lines in studies on co-infection of different pathogens and their interaction with the tick microbiome. We also carried out a preliminary study to investigate the effects of co-culturing Borrelia burgdorferi and Anaplasma phagocytophilum on their growth and interactions with the Ixodes ricinus cell line IRE/CTVM19 over a 13-day period. Replication of both pathogens was quantified by real-time PCR. The presence of A. phagocytophilum appeared to have a slight inhibitory effect on the multiplication of B. burgdorferi, that were added subsequently. In contrast, the prior presence of B. burgdorferi appeared to have a stimulatory effect on A. phagocytophilum after 6 days in culture. We conclude that the IRE/CTVM19 tick cell line is suitable for simultaneous and continuous cultivation of both bacteria and can be applied in future research.
Collapse
Affiliation(s)
- Violetta Zając
- Department of Health Biohazards and Parasitology, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland;
| | - Lesley Bell-Sakyi
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, 146 Brownlow Hill, Liverpool L3 5RF, UK;
| | - Angelina Wójcik-Fatla
- Department of Health Biohazards and Parasitology, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland;
| |
Collapse
|
5
|
Kumar D, Budachetri K, Rikihisa Y, Karim S. Analysis of Amblyomma americanum microRNAs in response to Ehrlichia chaffeensis infection and their potential role in vectorial capacity. Front Cell Infect Microbiol 2024; 14:1427562. [PMID: 39086604 PMCID: PMC11288922 DOI: 10.3389/fcimb.2024.1427562] [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: 05/04/2024] [Accepted: 06/27/2024] [Indexed: 08/02/2024] Open
Abstract
Background MicroRNAs (miRNAs) represent a subset of small noncoding RNAs and carry tremendous potential for regulating gene expression at the post-transcriptional level. They play pivotal roles in distinct cellular mechanisms including inhibition of bacterial, parasitic, and viral infections via immune response pathways. Intriguingly, pathogens have developed strategies to manipulate the host's miRNA profile, fostering environments conducive to successful infection. Therefore, changes in an arthropod host's miRNA profile in response to pathogen invasion could be critical in understanding host-pathogen dynamics. Additionally, this area of study could provide insights into discovering new targets for disease control and prevention. The main objective of the present study is to investigate the functional role of differentially expressed miRNAs upon Ehrlichia chaffeensis, a tick-borne pathogen, infection in tick vector, Amblyomma americanum. Methods Small RNA libraries from uninfected and E. chaffeensis-infected Am. americanum midgut and salivary gland tissues were prepared using the Illumina Truseq kit. Small RNA sequencing data was analyzed using miRDeep2 and sRNAtoolbox to identify novel and known miRNAs. The differentially expressed miRNAs were validated using a quantitative PCR assay. Furthermore, a miRNA inhibitor approach was used to determine the functional role of selected miRNA candidates. Results The sequencing of small RNA libraries generated >147 million raw reads in all four libraries and identified a total of >250 miRNAs across the four libraries. We identified 23 and 14 differentially expressed miRNAs in salivary glands, and midgut tissues infected with E. chaffeensis, respectively. Three differentially expressed miRNAs (miR-87, miR-750, and miR-275) were further characterized to determine their roles in pathogen infection. Inhibition of target miRNAs significantly decreased the E. chaffeensis load in tick tissues, which warrants more in-depth mechanistic studies. Conclusions The current study identified known and novel miRNAs and suggests that interfering with these miRNAs may impact the vectorial capacity of ticks to harbor Ehrlichia. This study identified several new miRNAs for future analysis of their functions in tick biology and tick-pathogen interaction studies.
Collapse
Affiliation(s)
- Deepak Kumar
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Khemraj Budachetri
- Laboratory of Molecular, Cellular, and Environmental Rickettsiology, Department of Veterinary Biosciences, College of Veterinary Medicine, Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States
| | - Yasuko Rikihisa
- Laboratory of Molecular, Cellular, and Environmental Rickettsiology, Department of Veterinary Biosciences, College of Veterinary Medicine, Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States
| | - Shahid Karim
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| |
Collapse
|
6
|
Kumar D, Budachetri K, Rikihisa Y, Karim S. Analysis of Amblyomma americanum microRNAs in response to Ehrlichia chaffeensis infection and their potential role in vectorial capacity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592465. [PMID: 38765993 PMCID: PMC11100627 DOI: 10.1101/2024.05.03.592465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Background MicroRNAs (miRNAs) represent a subset of small noncoding RNAs and carry tremendous potential for regulating gene expression at the post-transcriptional level. They play pivotal roles in distinct cellular mechanisms including inhibition of bacterial, parasitic, and viral infections via immune response pathways. Intriguingly, pathogens have developed strategies to manipulate the host's miRNA profile, fostering environments conducive to successful infection. Therefore, changes in an arthropod host's miRNA profile in response to pathogen invasion could be critical in understanding host-pathogen dynamics. Additionally, this area of study could provide insights into discovering new targets for disease control and prevention. The main objective of the present study is to investigate the functional role of differentially expressed miRNAs upon Ehrlichia chaffeensis, a tick-borne pathogen, infection in tick vector, Amblyomma americanum. Methods Small RNA libraries from uninfected and E. chaffeensis-infected Am. americanum midgut and salivary gland tissues were prepared using the Illumina Truseq kit. Small RNA sequencing data was analyzed using miRDeep2 and sRNAtoolbox to identify novel and known miRNAs. The differentially expressed miRNAs were validated using a quantitative PCR assay. Furthermore, a miRNA inhibitor approach was used to determine the functional role of selected miRNA candidates. Results The sequencing of small RNA libraries generated >147 million raw reads in all four libraries and identified a total of >250 miRNAs across the four libraries. We identified 23 and 14 differentially expressed miRNAs in salivary glands, and midgut tissues infected with E. chaffeensis, respectively. Three differentially expressed miRNAs (miR-87, miR-750, and miR-275) were further characterized to determine their roles in pathogen infection. Inhibition of target miRNAs significantly decreased the E. chaffeensis load in tick tissues, which warrants more in-depth mechanistic studies. Conclusions The current study identified known and novel miRNAs and suggests that interfering with these miRNAs may impact the vectorial capacity of ticks to harbor Ehrlichia. This study identified several new miRNAs for future analysis of their functions in tick biology and tick-pathogen interaction studies.
Collapse
Affiliation(s)
- Deepak Kumar
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Khemraj Budachetri
- Laboratory of Molecular, Cellular, and Environmental Rickettsiology, Department of Veterinary Biosciences, College of Veterinary Medicine, Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States
| | - Yasuko Rikihisa
- Laboratory of Molecular, Cellular, and Environmental Rickettsiology, Department of Veterinary Biosciences, College of Veterinary Medicine, Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States
| | - Shahid Karim
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS 39406, USA
| |
Collapse
|
7
|
Namjoshi P, Lubembe DM, Sultana H, Neelakanta G. Antibody-blocking of a tick transporter impairs Anaplasma phagocytophilum colonization in Haemaphysalis longicornis ticks. Sci Rep 2024; 14:9003. [PMID: 38637614 PMCID: PMC11026487 DOI: 10.1038/s41598-024-59315-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 04/09/2024] [Indexed: 04/20/2024] Open
Abstract
The invasive Asian longhorned tick Haemaphysalis longicornis that vectors and transmits several animal pathogens is significantly expanding in the United States. Recent studies report that these ticks also harbor human pathogens including Borrelia burgdorferi sensu lato, Babesia microti, and Anaplasma phagocytophilum. Therefore, studies that address the interactions of these ticks with human pathogens are important. In this study, we report the characterization of H. longicornis organic anion-transporting polypeptides (OATPs) in interactions of these ticks with A. phagocytophilum. Using OATP-signature sequence, we identified six OATPs in the H. longicornis genome. Bioinformatic analysis revealed that H. longicornis OATPs are closer to other tick orthologs rather than to mammalian counterparts. Quantitative real-time PCR analysis revealed that OATPs are highly expressed in immature stages when compared to mature stages of these ticks. In addition, we noted that the presence of A. phagocytophilum upregulates a specific OATP in these ticks. We also noted that exogenous treatment of H. longicornis with xanthurenic acid, a tryptophan metabolite, influenced OATP expression in these ticks. Immunoblotting analysis revealed that antibody generated against Ixodes scapularis OATP cross-reacted with H. longicornis OATP. Furthermore, treatment of H. longicornis with OATP antibody impaired colonization of A. phagocytophilum in these ticks. These results not only provide evidence that the OATP-tryptophan pathway is important for A. phagocytophilum survival in H. longicornis ticks but also indicate OATP as a promising candidate for the development of a universal anti-tick vaccine to target this bacterium and perhaps other rickettsial pathogens of medical importance.
Collapse
Affiliation(s)
- Prachi Namjoshi
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, 37996, USA
| | - Donald M Lubembe
- Department of Veterinary Pathology, Microbiology and Parasitology, Faculty of Veterinary Medicine and Surgery, Egerton University, Egerton, Kenya
| | - Hameeda Sultana
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, 37996, USA
| | - Girish Neelakanta
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, 37996, USA.
| |
Collapse
|
8
|
Liu SS, Fang X, Wen X, Liu JS, Alip M, Sun T, Wang YY, Chen HW. How mesenchymal stem cells transform into adipocytes: Overview of the current understanding of adipogenic differentiation. World J Stem Cells 2024; 16:245-256. [PMID: 38577237 PMCID: PMC10989283 DOI: 10.4252/wjsc.v16.i3.245] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/15/2024] [Accepted: 02/18/2024] [Indexed: 03/25/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are stem/progenitor cells capable of self-renewal and differentiation into osteoblasts, chondrocytes and adipocytes. The transformation of multipotent MSCs to adipocytes mainly involves two subsequent steps from MSCs to preadipocytes and further preadipocytes into adipocytes, in which the process MSCs are precisely controlled to commit to the adipogenic lineage and then mature into adipocytes. Previous studies have shown that the master transcription factors C/enhancer-binding protein alpha and peroxisome proliferation activator receptor gamma play vital roles in adipogenesis. However, the mechanism underlying the adipogenic differentiation of MSCs is not fully understood. Here, the current knowledge of adipogenic differentiation in MSCs is reviewed, focusing on signaling pathways, noncoding RNAs and epigenetic effects on DNA methylation and acetylation during MSC differentiation. Finally, the relationship between maladipogenic differentiation and diseases is briefly discussed. We hope that this review can broaden and deepen our understanding of how MSCs turn into adipocytes.
Collapse
Affiliation(s)
- Shan-Shan Liu
- Department of Reumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Xiang Fang
- Department of Emergency, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Xin Wen
- Department of Reumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Ji-Shan Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Miribangvl Alip
- Department of Reumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Tian Sun
- Department of Reumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu Province, China
| | - Yuan-Yuan Wang
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu 233000, Anhui Province, China
| | - Hong-Wei Chen
- Department of Reumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, Jiangsu Province, China.
| |
Collapse
|
9
|
Park JM, Genera BM, Fahy D, Swallow KT, Nelson CM, Oliver JD, Shaw DK, Munderloh UG, Brayton KA. An Anaplasma phagocytophilum T4SS effector, AteA, is essential for tick infection. mBio 2023; 14:e0171123. [PMID: 37747883 PMCID: PMC10653876 DOI: 10.1128/mbio.01711-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 07/26/2023] [Indexed: 09/27/2023] Open
Abstract
IMPORTANCE Ticks are the number one vector of pathogens for livestock worldwide and for humans in the United States. The biology of tick transmission is an understudied area. Understanding this critical interaction could provide opportunities to affect the course of disease spread. In this study, we examined the zoonotic tick-borne agent Anaplasma phagocytophilum and identified a secreted protein, AteA, which is expressed in a tick-specific manner. These secreted proteins, termed effectors, are the first proteins to interact with the host environment. AteA is essential for survival in ticks and appears to interact with cortical actin. Most effector proteins are studied in the context of the mammalian host; however, understanding how this unique set of proteins affects tick transmission is critical to developing interventions.
Collapse
Affiliation(s)
- Jason M. Park
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Brittany M. Genera
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Deirdre Fahy
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Kyle T. Swallow
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Curtis M. Nelson
- Department of Entomology, College of Food, Agricultural, and Natural Resource Sciences, University of Minnesota, Saint Paul, Minnesota, USA
| | - Jonathan D. Oliver
- Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Dana K. Shaw
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Ulrike G. Munderloh
- Department of Entomology, College of Food, Agricultural, and Natural Resource Sciences, University of Minnesota, Saint Paul, Minnesota, USA
| | - Kelly A. Brayton
- Program in Vector-borne Disease, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| |
Collapse
|
10
|
Mahesh PP, Namjoshi P, Sultana H, Neelakanta G. Immunization against arthropod protein impairs transmission of rickettsial pathogen from ticks to the vertebrate host. NPJ Vaccines 2023; 8:79. [PMID: 37253745 PMCID: PMC10229574 DOI: 10.1038/s41541-023-00678-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 05/16/2023] [Indexed: 06/01/2023] Open
Abstract
Human anaplasmosis caused by Anaplasma phagocytophilum is one of the most common tick-borne diseases in the United States. The black-legged ticks, Ixodes scapularis, vector and transmit this bacterium to humans. In this study, we provide evidence that targeting I. scapularis membrane-bound organic anion transporting polypeptide 4056 (IsOATP4056) with an anti-vector vaccine affects transmission of A. phagocytophilum from ticks to the vertebrate host. Anaplasma phagocytophilum induces expression of IsOATP4056 in ticks and tick cells. Increased membrane localization of IsOATP4056 was evident in A. phagocytophilum-infected tick cells. Treatment with high dose (10 µg/ml) but not low dose (5 µg/ml) of EL-6 antibody that targets the largest extracellular loop of IsOATP4056 showed cytotoxic effects in tick cells but not in human keratinocyte cell line (HaCaT). Passive immunization, tick-mediated transmission and in vitro studies performed with mice ordered from two commercial vendors and with tick cells showed that EL-6 antibody not only impairs A. phagocytophilum transmission from ticks to the murine host but also aids in the reduction in the bacterial loads within engorged ticks and in tick cells by activation of arthropod Toll pathway. Furthermore, reduced molting efficiency was noted in ticks fed on EL-6 antibody-immunized mice. Collectively, these results provide a good candidate for the development of anti-tick vaccine to target the transmission of A. phagocytophilum and perhaps other tick-borne pathogens of medical importance.
Collapse
Affiliation(s)
- P P Mahesh
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| | - Prachi Namjoshi
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| | - Hameeda Sultana
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA
| | - Girish Neelakanta
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, USA.
| |
Collapse
|
11
|
Tang H, Zhang D, Jiang F, Yu L, Tang H, Zhu J, Wu S, Niu H. Enhancement of Cell Adhesion by Anaplasma phagocytophilum Nucleolin-Interacting Protein AFAP. J Pers Med 2023; 13:jpm13020302. [PMID: 36836536 PMCID: PMC9965380 DOI: 10.3390/jpm13020302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Anaplasma phagocytophilum, the aetiologic agent of human granulocytic anaplasmosis (HGA), is an obligate intracellular Gram-negative bacterium. During infection, A. phagocytophilum enhances the adhesion of neutrophils to the infected endothelial cells. However, the bacterial factors contributing to this phenomenon remain unknown. In this study, we characterized a type IV secretion system substrate of A. phagocytophilum, AFAP (an actin filament-associated Anaplasma phagocytophilum protein) and found that it dynamically changed its pattern and subcellular location in cells and enhanced cell adhesion. Tandem affinity purification combined with mass spectrometry identified host nucleolin as an AFAP-interacting protein. Further study showed the disruption of nucleolin by RNA interference, and the treatment of a nucleolin-binding DNA aptamer AS1411 attenuated AFAP-mediated cell adhesion, indicating that AFAP enhanced cell adhesion in a nucleolin-dependent manner. The characterization of cell adhesion-enhancing AFAP and the identification of host nucleolin as its interaction partner may help understand the mechanism underlying A. phagocytophilum-promoting cell adhesion, facilitating the elucidation of HGA pathogenesis.
Collapse
Affiliation(s)
- Hongcheng Tang
- Department of Microbiology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Daxiu Zhang
- Clinical Laboratory Center, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Fenfen Jiang
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Lifeng Yu
- Clinical Laboratory Center, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Hui Tang
- Clinical Laboratory Center, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Jiafeng Zhu
- Department of Microbiology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Shuyan Wu
- Department of Microbiology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Hua Niu
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin 541001, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
- Correspondence:
| |
Collapse
|
12
|
Park JM, Genera BM, Fahy D, Swallow KT, Nelson CM, Oliver JD, Shaw DK, Munderloh UG, Brayton KA. An Anaplasma phagocytophilum T4SS effector, AteA, is essential for tick infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.06.527355. [PMID: 36798287 PMCID: PMC9934581 DOI: 10.1101/2023.02.06.527355] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Pathogens must adapt to disparate environments in permissive host species, a feat that is especially pronounced for vector-borne microbes, which transition between vertebrate hosts and arthropod vectors to complete their lifecycles. Most knowledge about arthropod-vectored bacterial pathogens centers on their life in the mammalian host, where disease occurs. However, disease outbreaks are driven by the arthropod vectors. Adapting to the arthropod is critical for obligate intracellular rickettsial pathogens, as they depend on eukaryotic cells for survival. To manipulate the intracellular environment, these bacteria use Type IV Secretion Systems (T4SS) to deliver effectors into the host cell. To date, few rickettsial T4SS translocated effectors have been identified and have only been examined in the context of mammalian infection. We identified an effector from the tick-borne rickettsial pathogen Anaplasma phagocytophilum , HGE1_02492, as critical for survival in tick cells and acquisition by ticks in vivo . Conversely, HGE1_02492 was dispensable during mammalian cell culture and murine infection. We show HGE1_02492 is translocatable in a T4SS-dependent manner to the host cell cytosol. In eukaryotic cells, the HGE1_02492 localized with cortical actin filaments, which is dependent on multiple sub-domains of the protein. HGE1_02492 is the first arthropod-vector specific T4SS translocated effector identified from a rickettsial pathogen. Moreover, the subcellular target of HGE1_02492 suggests that A. phagocytophilum is manipulating actin to enable arthropod colonization. Based on these findings, we propose the name AteA for Anaplasma ( phagocytophilum ) tick effector A. Altogether, we show that A. phagocytophilum uses distinct strategies to cycle between mammals and arthropods. Importance Ticks are the number one vector of pathogens for livestock worldwide and for humans in the US. The biology of tick transmission is an understudied area. Understanding this critical interaction could provide opportunities to affect the course of disease spread. In this study we examined the zoonotic tick-borne agent Anaplasma phagocytophilum and identified a secreted protein, AteA, that is expressed in a tick-specific manner. These secreted proteins, termed effectors, are the first proteins to interact with the host environment. AteA is essential for survival in ticks and appears to interact with cortical actin. Most effector proteins are studied in the context of the mammalian host; however, understanding how this unique set of proteins affect tick transmission is critical to developing interventions.
Collapse
|
13
|
Rickettsial pathogen inhibits tick cell death through tryptophan metabolite mediated activation of p38 MAP kinase. iScience 2022; 26:105730. [PMID: 36582833 PMCID: PMC9792911 DOI: 10.1016/j.isci.2022.105730] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/27/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Anaplasma phagocytophilum modulates various cell signaling pathways in mammalian cells for its survival. In this study, we report that A. phagocytophilum modulates tick tryptophan pathway to activate arthropod p38 MAP kinase for the survival of both this bacterium and its vector host. Increased level of tryptophan metabolite, xanthurenic acid (XA), was evident in A. phagocytophilum-infected ticks and tick cells. Lower levels of cell death markers and increased levels of total and phosphorylated p38 MAPK was noted in A. phagocytophilum-infected ticks and tick cells. Treatment with XA increased phosphorylated p38 MAPK levels and reduced cell death in A. phagocytophilum-infected tick cells. Furthermore, treatment with p38 MAPK inhibitor affected bacterial replication, decreased phosphorylated p38 MAPK levels and increased tick cell death. However, XA reversed these effects. Taken together, we provide evidence that rickettsial pathogen modulates arthropod tryptophan and p38 MAPK pathways to inhibit cell death for its survival in ticks.
Collapse
|
14
|
Kumar D, Downs LP, Embers M, Flynt AS, Karim S. Identification of microRNAs in the Lyme Disease Vector Ixodes scapularis. Int J Mol Sci 2022; 23:5565. [PMID: 35628370 PMCID: PMC9141961 DOI: 10.3390/ijms23105565] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 02/01/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs involved in many biological processes, including the immune pathways that control bacterial, parasitic, and viral infections. Pathogens probably modify host miRNAs to facilitate successful infection, so they might be useful targets for vaccination strategies. There are few data on differentially expressed miRNAs in the black-legged tick Ixodes scapularis after infection with Borrelia burgdorferi, the causative agent of Lyme disease in the United States. Small RNA sequencing and qRT-PCR analysis were used to identify and validate differentially expressed I. scapularis salivary miRNAs. Small RNA-seq yielded 133,465,828 (≥18 nucleotides) and 163,852,135 (≥18 nucleotides) small RNA reads from Borrelia-infected and uninfected salivary glands for downstream analysis using the miRDeep2 algorithm. As such, 254 miRNAs were identified across all datasets, 25 of which were high confidence and 51 low confidence known miRNAs. Further, 23 miRNAs were differentially expressed in uninfected and infected salivary glands: 11 were upregulated and 12 were downregulated upon pathogen infection. Gene ontology and network analysis of target genes of differentially expressed miRNAs predicted roles in metabolic, cellular, development, cellular component biogenesis, and biological regulation processes. Several Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, including sphingolipid metabolism; valine, leucine and isoleucine degradation; lipid transport and metabolism; exosome biogenesis and secretion; and phosphate-containing compound metabolic processes, were predicted as targets of differentially expressed miRNAs. A qRT-PCR assay was utilized to validate the differential expression of miRNAs. This study provides new insights into the miRNAs expressed in I. scapularis salivary glands and paves the way for their functional manipulation to prevent or treat B. burgdorferi infection.
Collapse
Affiliation(s)
- Deepak Kumar
- Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA; (D.K.); (A.S.F.)
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA;
| | - Latoyia P. Downs
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA;
| | - Monica Embers
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA;
| | - Alex Sutton Flynt
- Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA; (D.K.); (A.S.F.)
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA;
| | - Shahid Karim
- Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA; (D.K.); (A.S.F.)
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS 39406, USA;
| |
Collapse
|
15
|
Khanal S, Taank V, Anderson JF, Sultana H, Neelakanta G. Rickettsial Pathogen Perturbs Tick Circadian Gene to Infect the Vertebrate Host. Int J Mol Sci 2022; 23:3545. [PMID: 35408905 PMCID: PMC8998576 DOI: 10.3390/ijms23073545] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/14/2022] [Accepted: 03/19/2022] [Indexed: 11/20/2022] Open
Abstract
Ixodes scapularis is a medically important tick that transmits several microbes to humans, including rickettsial pathogen Anaplasma phagocytophilum. In nature, these ticks encounter several abiotic factors including changes in temperature, humidity, and light. Many organisms use endogenously generated circadian pathways to encounter abiotic factors. In this study, we provide evidence for the first time to show that A. phagocytophilum modulates the arthropod circadian gene for its transmission to the vertebrate host. We noted a circadian oscillation in the expression of arthropod clock, bmal1, period and timeless genes when ticks or tick cells were exposed to alternate 12 h light: 12 h dark conditions. Moreover, A. phagocytophilum significantly modulates the oscillation pattern of expression of these genes. In addition, increased levels of clock and bmal1 and decreased expression of Toll and JAK/STAT pathway immune genes such as pelle and jak, respectively, were noted during A. phagocytophilum transmission from ticks to the vertebrate host. RNAi-mediated knockdown of clock gene expression in ticks resulted in the reduced expression of jak and pelle that increased bacterial transmission from ticks to the murine host. Furthermore, clock-deficient ticks fed late and had less engorgement weights. These results indicate an important role for circadian modulation of tick gene expression that is critical for arthropod blood feeding and transmission of pathogens from vector to the vertebrate host.
Collapse
Affiliation(s)
- Supreet Khanal
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA; (S.K.); (V.T.); (H.S.)
| | - Vikas Taank
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA; (S.K.); (V.T.); (H.S.)
| | - John F. Anderson
- Department of Entomology, Connecticut Agricultural Experiment Station, New Haven, CT 06511, USA;
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA; (S.K.); (V.T.); (H.S.)
- Center for Molecular Medicine, Old Dominion University, Norfolk, VA 23529, USA
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA; (S.K.); (V.T.); (H.S.)
- Center for Molecular Medicine, Old Dominion University, Norfolk, VA 23529, USA
| |
Collapse
|
16
|
Hromníková D, Furka D, Furka S, Santana JAD, Ravingerová T, Klöcklerová V, Žitňan D. Prevention of tick-borne diseases: challenge to recent medicine. Biologia (Bratisl) 2022; 77:1533-1554. [PMID: 35283489 PMCID: PMC8905283 DOI: 10.1007/s11756-021-00966-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 11/10/2021] [Indexed: 12/14/2022]
Abstract
Abstract Ticks represent important vectors and reservoirs of pathogens, causing a number of diseases in humans and animals, and significant damage to livestock every year. Modern research into protection against ticks and tick-borne diseases focuses mainly on the feeding stage, i.e. the period when ticks take their blood meal from their hosts during which pathogens are transmitted. Physiological functions in ticks, such as food intake, saliva production, reproduction, development, and others are under control of neuropeptides and peptide hormones which may be involved in pathogen transmission that cause Lyme borreliosis or tick-borne encephalitis. According to current knowledge, ticks are not reservoirs or vectors for the spread of COVID-19 disease. The search for new vaccination methods to protect against ticks and their transmissible pathogens is a challenge for current science in view of global changes, including the increasing migration of the human population. Highlights • Tick-borne diseases have an increasing incidence due to climate change and increased human migration • To date, there is no evidence of transmission of coronavirus COVID-19 by tick as a vector • To date, there are only a few modern, effective, and actively- used vaccines against ticks or tick-borne diseases • Neuropeptides and their receptors expressed in ticks may be potentially used for vaccine design
Collapse
Affiliation(s)
- Dominika Hromníková
- Department of Molecular Physiology, Slovak Academy of Sciences, Institute of Zoology, Dúbravská cesta 9, 84506 Bratislava, Slovakia
| | - Daniel Furka
- Faculty of Natural Sciences, Department of Physical and Theoretical Chemistry, Comenius University, Mlynská dolina, Ilkovičova 6, 84104 Bratislava, SK Slovakia
- Department of Cardiovascular Physiology and Pathophysiology, Slovak Academy of Sciences, Institute of Heart Research, Dúbravská cesta 9, SK 84005 Bratislava, Slovakia
| | - Samuel Furka
- Faculty of Natural Sciences, Department of Physical and Theoretical Chemistry, Comenius University, Mlynská dolina, Ilkovičova 6, 84104 Bratislava, SK Slovakia
- Department of Cardiovascular Physiology and Pathophysiology, Slovak Academy of Sciences, Institute of Heart Research, Dúbravská cesta 9, SK 84005 Bratislava, Slovakia
| | - Julio Ariel Dueñas Santana
- Chemical Engineering Department, University of Matanzas, Km 3 Carretera a Varadero, 44740 Matanzas, CU Cuba
| | - Táňa Ravingerová
- Department of Cardiovascular Physiology and Pathophysiology, Slovak Academy of Sciences, Institute of Heart Research, Dúbravská cesta 9, SK 84005 Bratislava, Slovakia
| | - Vanda Klöcklerová
- Department of Molecular Physiology, Slovak Academy of Sciences, Institute of Zoology, Dúbravská cesta 9, 84506 Bratislava, Slovakia
| | - Dušan Žitňan
- Department of Molecular Physiology, Slovak Academy of Sciences, Institute of Zoology, Dúbravská cesta 9, 84506 Bratislava, Slovakia
| |
Collapse
|
17
|
Neelakanta G, Sultana H. Tick Saliva and Salivary Glands: What Do We Know So Far on Their Role in Arthropod Blood Feeding and Pathogen Transmission. Front Cell Infect Microbiol 2022; 11:816547. [PMID: 35127563 PMCID: PMC8809362 DOI: 10.3389/fcimb.2021.816547] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/22/2021] [Indexed: 12/25/2022] Open
Abstract
Ticks are blood-sucking arthropods that have developed myriad of strategies to get a blood meal from the vertebrate host. They first attach to the host skin, select a bite site for a blood meal, create a feeding niche at the bite site, secrete plethora of molecules in its saliva and then starts feeding. On the other side, host defenses will try to counter-attack and stop tick feeding at the bite site. In this constant battle between ticks and the host, arthropods successfully pacify the host and completes a blood meal and then replete after full engorgement. In this review, we discuss some of the known and emerging roles for arthropod components such as cement, salivary proteins, lipocalins, HSP70s, OATPs, and extracellular vesicles/exosomes in facilitating successful blood feeding from ticks. In addition, we discuss how tick-borne pathogens modulate(s) these components to infect the vertebrate host. Understanding the biology of arthropod blood feeding and molecular interactions at the tick-host interface during pathogen transmission is very important. This information would eventually lead us in the identification of candidates for the development of transmission-blocking vaccines to prevent diseases caused by medically important vector-borne pathogens.
Collapse
|
18
|
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
|
19
|
Taank V, Lattanzio FA, Sultana H, Neelakanta G. Double anus in an Ixodes scapularis nymph, a medically important tick vector. Parasit Vectors 2021; 14:251. [PMID: 33975641 PMCID: PMC8112061 DOI: 10.1186/s13071-021-04757-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/29/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ixodes scapularis ticks are medically important arthropod vectors that transmit several pathogens to humans. The observations of morphological abnormalities, including nanism, missing leg, extra leg, and gynandromorphism, have been reported in these ticks. In this study, we report the presence of two anuses in a laboratory-reared I. scapularis nymph. RESULTS Larval ticks were allowed to feed on mice and to molt to nymphs. Two anuses were observed in one of the freshly molted nymphs. Stereo and scanning electron microscopy confirmed the presence of two anuses in one nymph within a single anal groove. CONCLUSIONS This report confirms the rare occurrence of double anus in I. scapularis.
Collapse
Affiliation(s)
- Vikas Taank
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - Frank A Lattanzio
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA.,Center for Molecular Medicine, Old Dominion University, Norfolk, VA, USA.,Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, 37996, USA
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA. .,Center for Molecular Medicine, Old Dominion University, Norfolk, VA, USA. .,Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, 37996, USA.
| |
Collapse
|
20
|
O'Neal AJ, Singh N, Mendes MT, Pedra JHF. The genus Anaplasma: drawing back the curtain on tick-pathogen interactions. Pathog Dis 2021; 79:ftab022. [PMID: 33792663 PMCID: PMC8062235 DOI: 10.1093/femspd/ftab022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022] Open
Abstract
Tick-borne illnesses pose a serious concern to human and veterinary health and their prevalence is on the rise. The interactions between ticks and the pathogens they carry are largely undefined. However, the genus Anaplasma, a group of tick-borne bacteria, has been instrumental in uncovering novel paradigms in tick biology. The emergence of sophisticated technologies and the convergence of entomology with microbiology, immunology, metabolism and systems biology has brought tick-Anaplasma interactions to the forefront of vector biology with broader implications for the infectious disease community. Here, we discuss the use of Anaplasma as an instrument for the elucidation of novel principles in arthropod-microbe interactions. We offer an outlook of the primary areas of study, outstanding questions and future research directions.
Collapse
Affiliation(s)
- Anya J O'Neal
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Nisha Singh
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Maria Tays Mendes
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
21
|
Taank V, Ramasamy E, Sultana H, Neelakanta G. An efficient microinjection method to generate human anaplasmosis agent Anaplasma phagocytophilum-infected ticks. Sci Rep 2020; 10:15994. [PMID: 32994497 PMCID: PMC7524789 DOI: 10.1038/s41598-020-73061-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 09/03/2020] [Indexed: 11/09/2022] Open
Abstract
Ticks are important vectors that transmit several pathogens including human anaplasmosis agent, Anaplasma phagocytophilum. This bacterium is an obligate intracellular rickettsial pathogen. An infected reservoir animal host is often required for maintenance of this bacterial colony and as a source for blood to perform needle inoculations in naïve animals for tick feeding studies. In this study, we report an efficient microinjection method to generate A. phagocytophilum-infected ticks in laboratory conditions. The dense-core (DC) form of A. phagocytophilum was isolated from in vitro cultures and injected into the anal pore of unfed uninfected Ixodes scapularis nymphal ticks. These ticks successfully transmitted A. phagocytophilum to the murine host. The bacterial loads were detected in murine blood, spleen, and liver tissues. In addition, larval ticks successfully acquired A. phagocytophilum from mice that were previously infected by feeding with DC-microinjected nymphal ticks. Transstadial transmission of A. phagocytophilum from larvae to nymphal stage was also evident in these ticks. Taken together, our study provides a timely, rapid, and an efficient method not only to generate A. phagocytophilum-infected ticks but also provides a tool to understand acquisition and transmission dynamics of this bacterium and perhaps other rickettsial pathogens from medically important vectors.
Collapse
Affiliation(s)
- Vikas Taank
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - Ellango Ramasamy
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, 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. .,Department of Biological Sciences, Center for Molecular Medicine, College of Sciences, Old Dominion University, Norfolk, VA, 23529, USA.
| |
Collapse
|
22
|
Jia N, Wang J, Shi W, Du L, Sun Y, Zhan W, Jiang JF, Wang Q, Zhang B, Ji P, Bell-Sakyi L, Cui XM, Yuan TT, Jiang BG, Yang WF, Lam TTY, Chang QC, Ding SJ, Wang XJ, Zhu JG, Ruan XD, Zhao L, Wei JT, Ye RZ, Que TC, Du CH, Zhou YH, Cheng JX, Dai PF, Guo WB, Han XH, Huang EJ, Li LF, Wei W, Gao YC, Liu JZ, Shao HZ, Wang X, Wang CC, Yang TC, Huo QB, Li W, Chen HY, Chen SE, Zhou LG, Ni XB, Tian JH, Sheng Y, Liu T, Pan YS, Xia LY, Li J, Zhao F, Cao WC. Large-Scale Comparative Analyses of Tick Genomes Elucidate Their Genetic Diversity and Vector Capacities. Cell 2020; 182:1328-1340.e13. [PMID: 32814014 DOI: 10.1016/j.cell.2020.07.023] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/01/2020] [Accepted: 07/17/2020] [Indexed: 12/12/2022]
Abstract
Among arthropod vectors, ticks transmit the most diverse human and animal pathogens, leading to an increasing number of new challenges worldwide. Here we sequenced and assembled high-quality genomes of six ixodid tick species and further resequenced 678 tick specimens to understand three key aspects of ticks: genetic diversity, population structure, and pathogen distribution. We explored the genetic basis common to ticks, including heme and hemoglobin digestion, iron metabolism, and reactive oxygen species, and unveiled for the first time that genetic structure and pathogen composition in different tick species are mainly shaped by ecological and geographic factors. We further identified species-specific determinants associated with different host ranges, life cycles, and distributions. The findings of this study are an invaluable resource for research and control of ticks and tick-borne diseases.
Collapse
Affiliation(s)
- Na Jia
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China; Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing 100071, P.R. China
| | - Jinfeng Wang
- Computational Genomics Lab, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, P.R. China; State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Wenqiang Shi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Lifeng Du
- Computational Genomics Lab, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, P.R. China; Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan 250012, Shandong, P.R. China
| | - Yi Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Wei Zhan
- Annoroad Gene Technology (Beijing) Company Limited, Beijing 100176, P.R. China
| | - Jia-Fu Jiang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China; Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing 100071, P.R. China
| | - Qian Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China; Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan 250012, Shandong, P.R. China
| | - Bing Zhang
- Computational Genomics Lab, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Peifeng Ji
- Computational Genomics Lab, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Lesley Bell-Sakyi
- Department of Infection Biology and Microbiomes, Institute of Infection, Ecological and Veterinary Sciences, University of Liverpool, Liverpool L3 5RF, UK
| | - Xiao-Ming Cui
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China; Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing 100071, P.R. China
| | - Ting-Ting Yuan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Bao-Gui Jiang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Wei-Fei Yang
- Annoroad Gene Technology (Beijing) Company Limited, Beijing 100176, P.R. China
| | - Tommy Tsan-Yuk Lam
- State Key Laboratory of Emerging Infectious Diseases and Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Qiao-Cheng Chang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang, P.R. China
| | - Shu-Jun Ding
- Shandong Center for Disease Control and Prevention, Shandong Provincial Key Laboratory of Communicable Disease Control and Prevention, Jinan 250014, Shandong, P.R. China
| | - Xian-Jun Wang
- Shandong Center for Disease Control and Prevention, Shandong Provincial Key Laboratory of Communicable Disease Control and Prevention, Jinan 250014, Shandong, P.R. China
| | - Jin-Guo Zhu
- ManZhouLi Customs District, Manzhouli 021400, Inner Mongolia, P.R. China
| | - Xiang-Dong Ruan
- Academy of Forest Inventory and Planning, State Forestry and Grassland Administration, Beijing 100714, P.R. China
| | - Lin Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China; Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan 250012, Shandong, P.R. China
| | - Jia-Te Wei
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China; Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan 250012, Shandong, P.R. China
| | - Run-Ze Ye
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China; Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan 250012, Shandong, P.R. China
| | - Teng Cheng Que
- Guangxi Zhuang Autonomous Region Terrestrial Wildlife Medical-aid and Monitoring Epidemic Diseases Research Center, Nanjing 530028, Guangxi, P.R. China
| | - Chun-Hong Du
- Yunnan Institute for Endemic Diseases Control and Prevention, Dali 671000, Yunnan, P.R. China
| | - Yu-Hao Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Jing Xia Cheng
- Shanxi Provence Center for Disease Control and Prevention, Xian 030012, Shanxi, P.R. China
| | - Pei-Fang Dai
- Shanxi Provence Center for Disease Control and Prevention, Xian 030012, Shanxi, P.R. China
| | - Wen-Bin Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Xiao-Hu Han
- Shenyang Agriculture University, Shenyang 110866, Liaoning, P.R. China
| | - En-Jiong Huang
- Fuzhou International Travel Healthcare Center, Fuzhou 350001, Fujian, P.R. China
| | - Lian-Feng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Wei Wei
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Yu-Chi Gao
- Annoroad Gene Technology (Beijing) Company Limited, Beijing 100176, P.R. China
| | - Jing-Ze Liu
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, P.R. China
| | - Hong-Ze Shao
- Animal Husbandry and Veterinary Science Research Institute of Jilin Province, Changchun 130062, Jilin, P.R. China
| | - Xin Wang
- Qingjiangpu District Center for Disease Control and Prevention, Huai'an 223001, Jiangsu, P.R. China
| | - Chong-Cai Wang
- Hainan International Travel Healthcare Center, Haikou 570311, Hainan, P.R. China
| | - Tian-Ci Yang
- State Key Lab of Mosquito-borne Diseases, Hangzhou International Tourism Healthcare Center, Hangzhou Customs of China, Hangzhou 310012, Zhejiang, P.R. China
| | - Qiu-Bo Huo
- Mudanjiang Forestry Central Hospital, Mudanjiang 157000, Heilongjiang, P.R. China
| | - Wei Li
- Xinjiang Center for Disease Control and Prevention, Urumqi 830002, Xinjiang, P.R. China
| | - Hai-Ying Chen
- Collaboration Unit for Field Epidemiology of the State Key Laboratory for Infectious Disease Prevention and Control, Nanchang Center for Disease Control and Prevention. Nanchang 330038, Jiangxi, P.R. China
| | - Shen-En Chen
- Collaboration Unit for Field Epidemiology of the State Key Laboratory for Infectious Disease Prevention and Control, Nanchang Center for Disease Control and Prevention. Nanchang 330038, Jiangxi, P.R. China
| | - Ling-Guo Zhou
- Shaanxi Natural Reserve and Wildlife Administration Station, Xi'an 710082, Shaanxi, P.R. China
| | - Xue-Bing Ni
- State Key Laboratory of Emerging Infectious Diseases and Centre of Influenza Research, School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Jun-Hua Tian
- Wuhan Center for Disease Control and Prevention, Wuhan 430015, Hubei, P.R. China
| | - Yue Sheng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Tao Liu
- Annoroad Gene Technology (Beijing) Company Limited, Beijing 100176, P.R. China
| | - Yu-Sheng Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Luo-Yuan Xia
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Jie Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China
| | - Fangqing Zhao
- Computational Genomics Lab, Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, P.R. China; State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, P.R. China; Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing 100071, P.R. China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, Yunan, P.R. China; Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of the Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, Zhejiang, P.R. China; University of the Chinese Academy of Sciences, Beijing 100049, P.R. China.
| | - Wu-Chun Cao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, P.R. China; Institute of EcoHealth, School of Public Health, Shandong University, 44 Wenhuaxi Street, Jinan 250012, Shandong, P.R. China; Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing 100071, P.R. China.
| |
Collapse
|
23
|
Tang H, Zhu J, Wu S, Niu H. Identification and characterization of an actin filament-associated Anaplasma phagocytophilum protein. Microb Pathog 2020; 147:104439. [PMID: 32768516 DOI: 10.1016/j.micpath.2020.104439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 08/03/2020] [Indexed: 10/23/2022]
Abstract
Anaplasma phagocytophilum, the aetiologic agent of human granulocytic anaplasmosis (HGA) is an obligate intracellular Gram-negative bacterium. During intracellular replication, A. phagocytophilum interacts with many host cell components including actin cytoskeleton. However the bacterial factors contributing to the interaction between A. phagocytophilum and actin filaments remain unknown. In this study we identified a novel type IV secretion system substrate of A. phagocytophilum by employing TEM-1 β-lactamase based protein translocation assay, and found it is an actin filament-associated protein. Here, we name this protein as an actin filament-associated Anaplasma phagocytophilumprotein (AFAP). Further analysis showed that the middle region of AFAP harboring four tandem repeats is involved in its interaction with actin filaments. The identification and characterization of an actin filament-associated A. phagocytophilum protein in this study may help understand the interaction between A. phagocytophilum and actin cytoskeleton of its host cells, facilitating the elucidation of HGA pathogenesis.
Collapse
Affiliation(s)
- Hongcheng Tang
- Department of Microbiology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Jiafeng Zhu
- Department of Microbiology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Shuyan Wu
- Department of Microbiology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Hua Niu
- Department of Microbiology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, 215123, China.
| |
Collapse
|
24
|
Dahmani M, Anderson JF, Sultana H, Neelakanta G. Rickettsial pathogen uses arthropod tryptophan pathway metabolites to evade reactive oxygen species in tick cells. Cell Microbiol 2020; 22:e13237. [PMID: 32562372 DOI: 10.1111/cmi.13237] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/10/2020] [Accepted: 06/13/2020] [Indexed: 12/16/2022]
Abstract
Reactive oxygen species (ROS) that are induced upon pathogen infection plays an important role in host defence. The rickettsial pathogen Anaplasma phagocytophilum, which is primarily transmitted by Ixodes scapularis ticks in the United States, has evolved many strategies to escape ROS and survive in mammalian cells. However, little is known on the role of ROS in A. phagocytophilum infection in ticks. Our results show that A. phagocytophilum and hemin induce activation of l-tryptophan pathway in tick cells. Xanthurenic acid (XA), a tryptophan metabolite, supports A. phagocytophilum growth in tick cells through inhibition of tryptophan dioxygenase (TDO) activity leading to reduced l-kynurenine levels that subsequently affects build-up of ROS. However, hemin supports A. phagocytophilum growth in tick cells by inducing TDO activity leading to increased l-kynurenine levels and ROS production. Our data reveal that XA and kynurenic acid (KA) chelate hemin. Furthermore, treatment of tick cells with 3-hydroxyl l-kynurenine limits A. phagocytophilum growth in tick cells. RNAi-mediated knockdown of kynurenine aminotransferase expression results in increased ROS production and reduced A. phagocytophilum burden in tick cells. Collectively, these results suggest that l-tryptophan pathway metabolites influence A. phagocytophilum survival by affecting build up of ROS levels in tick cells.
Collapse
Affiliation(s)
- Mustapha Dahmani
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, USA.,Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - John F Anderson
- Department of Entomology, Connecticut Agricultural Experiment Station, New Haven, Connecticut, USA
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, USA.,Center for Molecular Medicine, Old Dominion University, Norfolk, VA, USA
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, USA.,Center for Molecular Medicine, Old Dominion University, Norfolk, VA, USA
| |
Collapse
|
25
|
Khan AU, Qu R, Fan T, Ouyang J, Dai J. A glance on the role of actin in osteogenic and adipogenic differentiation of mesenchymal stem cells. Stem Cell Res Ther 2020; 11:283. [PMID: 32678016 PMCID: PMC7364498 DOI: 10.1186/s13287-020-01789-2] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/13/2020] [Accepted: 06/23/2020] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have the capacity to differentiate into multiple lineages including osteogenic and adipogenic lineages. An increasing number of studies have indicated that lineage commitment by MSCs is influenced by actin remodeling. Moreover, actin has roles in determining cell shape, nuclear shape, cell spreading, and cell stiffness, which eventually affect cell differentiation. Osteogenic differentiation is promoted in MSCs that exhibit a large spreading area, increased matrix stiffness, higher levels of actin polymerization, and higher density of stress fibers, whereas adipogenic differentiation is prevalent in MSCs with disrupted actin networks. In addition, the mechanical properties of F-actin empower cells to sense and transduce mechanical stimuli, which are also reported to influence differentiation. Various biomaterials, mechanical, and chemical interventions along with pathogen-induced actin alteration in the form of polymerization and depolymerization in MSC differentiation were studied recently. This review will cover the role of actin and its modifications through the use of different methods in inducing osteogenic and adipogenic differentiation.
Collapse
Affiliation(s)
- Asmat Ullah Khan
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Rongmei Qu
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Tingyu Fan
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Zhou W, Tahir F, Wang JCY, Woodson M, Sherman MB, Karim S, Neelakanta G, Sultana H. Discovery of Exosomes From Tick Saliva and Salivary Glands Reveals Therapeutic Roles for CXCL12 and IL-8 in Wound Healing at the Tick-Human Skin Interface. Front Cell Dev Biol 2020; 8:554. [PMID: 32766239 PMCID: PMC7378379 DOI: 10.3389/fcell.2020.00554] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 06/10/2020] [Indexed: 12/30/2022] Open
Abstract
Ticks secrete various anti-coagulatory, anti-vasoconstrictory, anti-inflammatory, and anti-platelet aggregation factors in their saliva at the bite site during feeding to evade host immunological surveillance and responses. For the first time, we report successful isolation of exosomes (small membrane-bound extracellular signaling vesicles) from saliva and salivary glands of partially fed or unfed ixodid ticks. Our data showed a novel role of these in vivo exosomes in the inhibition of wound healing via downregulation of C-X-C motif chemokine ligand 12 (CXCL12) and upregulation of interleukin-8 (IL-8). Cryo-electron microscopy (cryo-EM) analysis revealed that tick saliva and salivary glands are composed of heterogeneous populations of in vivo exosomes with sizes ranging from 30 to 200 nm. Enriched amounts of tick CD63 ortholog protein and heat shock protein 70 (HSP70) were evident in these exosomes. Treatment of human skin keratinocytes (HaCaT cells) with exosomes derived from tick saliva/salivary glands or ISE6 cells dramatically delayed cell migration, wound healing, and repair process. Wound healing is a highly dynamic process with several individualized processes including secretion of cytokines. Cytokine array profiling followed by immunoblotting and quantitative-PCR analysis revealed that HaCaT cells treated with exosomes derived from tick saliva/salivary glands or ISE6 cells showed enhanced IL-8 levels and reduced CXCL12 loads. Inhibition of IL-8 or CXCL12 further delayed exosome-mediated cell migration, wound healing, and repair process, suggesting a skin barrier protection role for these chemokines at the tick bite site. In contrast, exogenous treatment of CXCL12 protein completely restored this delay and enhanced the repair process. Taken together, our study provides novel insights on how tick salivary exosomes secreted in saliva can delay wound healing at the bite site to facilitate successful blood feeding.
Collapse
Affiliation(s)
- Wenshuo Zhou
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States
| | - Faizan Tahir
- Center for Molecular and Cellular Biosciences, School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Joseph Che-Yen Wang
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, United States.,Electron Microscopy Center, Indiana University, Bloomington, IN, United States
| | - Michael Woodson
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Michael B Sherman
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, United States.,Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Shahid Karim
- Center for Molecular and Cellular Biosciences, School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States.,Center for Molecular Medicine, Old Dominion University, Norfolk, VA, United States
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States.,Center for Molecular Medicine, Old Dominion University, Norfolk, VA, United States.,Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
27
|
Ramasamy E, Taank V, Anderson JF, Sultana H, Neelakanta G. Repression of tick microRNA-133 induces organic anion transporting polypeptide expression critical for Anaplasma phagocytophilum survival in the vector and transmission to the vertebrate host. PLoS Genet 2020; 16:e1008856. [PMID: 32614824 PMCID: PMC7331985 DOI: 10.1371/journal.pgen.1008856] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 05/13/2020] [Indexed: 01/16/2023] Open
Abstract
The microRNAs (miRNAs) are important regulators of gene expression. In this study, we provide evidence for the first time to show that rickettsial pathogen Anaplasma phagocytophilum infection results in the down-regulation of tick microRNA-133 (miR-133), to induce Ixodes scapularis organic anion transporting polypeptide (isoatp4056) gene expression critical for this bacterial survival in the vector and for its transmission to the vertebrate host. Transfection studies with recombinant constructs containing transcriptional fusions confirmed binding of miR-133 to isoatp4056 mRNA. Treatment with miR-133 inhibitor resulted in increased bacterial burden and isoatp4056 expression in ticks and tick cells. In contrast, treatment with miR-133 mimic or pre-mir-133 resulted in dramatic reduction in isoatp4056 expression and bacterial burden in ticks and tick cells. Moreover, treatment of ticks with pre-mir-133 affected vector-mediated A. phagocytophilum infection of murine host. These results provide novel insights to understand impact of modulation of tick miRNAs on pathogen colonization in the vector and their transmission to infect the vertebrate host. This study provides novel evidence that shows that down-regulation of arthropod microRNA-133 leading to enhanced expression of organic anion transporting polypeptide is not only critical for rickettsial pathogen Anaplasma phagocytophilum survival in ticks but also for this bacterial transmission from vector to the vertebrate host. Understanding how pathogens manipulate vector-signaling repertoire for their benefit would lead to the development of strategies to block their transmission from vector to the vertebrate host.
Collapse
Affiliation(s)
- Ellango Ramasamy
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, United States of America
| | - Vikas Taank
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, United States of America
| | - John F Anderson
- Department of Entomology, Connecticut Agricultural Experiment Station, New Haven, Connecticut, United States of America
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, United States of America
- Center for Molecular Medicine, Old Dominion University, Norfolk, Virginia, United States of America
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, United States of America
- Center for Molecular Medicine, Old Dominion University, Norfolk, Virginia, United States of America
- * E-mail:
| |
Collapse
|
28
|
Regmi P, Khanal S, Neelakanta G, Sultana H. Tick-Borne Flavivirus Inhibits Sphingomyelinase ( IsSMase), a Venomous Spider Ortholog to Increase Sphingomyelin Lipid Levels for Its Survival in Ixodes scapularis Ticks. Front Cell Infect Microbiol 2020; 10:244. [PMID: 32656091 PMCID: PMC7325911 DOI: 10.3389/fcimb.2020.00244] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/27/2020] [Indexed: 12/24/2022] Open
Abstract
Our previous study showed that cells from medically important arthropods, such as ticks, secrete extracellular vesicles (EVs) including exosomes that mediate transmission of flavivirus RNA and proteins to the human cells. Understanding the molecular determinants and mechanism(s) of arthropod-borne flavivirus transmission via exosome biogenesis is very important. In this current study, we showed that in the presence of tick-borne Langat Virus (LGTV; a member of tick-borne encephalitis virus complex), the expression of arthropod IsSMase, a sphingomyelinase D (SMase D) that catalyzes the hydrolytic cleavage of substrates like sphingomyelin (SM) lipids, was significantly reduced in both Ixodes scapularis ticks (in vivo) and in tick cells (in vitro). The IsSMase reduced levels correlated with down-regulation of its activity upon LGTV replication in tick cells. Our data show that LGTV-mediated suppression of IsSMase allowed accumulation of SM lipid levels that supported membrane-associated viral replication and exosome biogenesis. Inhibition of viral loads and SM lipid built up upon GW4869 inhibitor treatment reversed the IsSMase levels and restored its activity. Our results suggest an important role for this spider venomous ortholog IsSMase in regulating viral replication associated with membrane-bound SM lipids in ticks. In summary, our study not only suggests a novel role for arthropod IsSMase in tick-LGTV interactions but also provides new insights into its important function in vector defense mechanism(s) against tick-borne virus infection and in anti-viral pathway(s).
Collapse
Affiliation(s)
- Pravesh Regmi
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States
| | - Supreet Khanal
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States.,Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
29
|
Bhowmick B, Han Q. Understanding Tick Biology and Its Implications in Anti-tick and Transmission Blocking Vaccines Against Tick-Borne Pathogens. Front Vet Sci 2020; 7:319. [PMID: 32582785 PMCID: PMC7297041 DOI: 10.3389/fvets.2020.00319] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 05/11/2020] [Indexed: 12/13/2022] Open
Abstract
Ticks are obligate blood-feeding ectoparasites that transmit a wide variety of pathogens to animals and humans in many parts of the world. Currently, tick control methods primarily rely on the application of chemical acaricides, which results in the development of resistance among tick populations and environmental contamination. Therefore, an alternative tick control method, such as vaccines have been shown to be a feasible strategy that offers a sustainable, safe, effective, and environment-friendly solution. Nevertheless, novel control methods are hindered by a lack of understanding of tick biology, tick-pathogen-host interface, and identification of effective antigens in the development of vaccines. This review highlights the current knowledge and data on some of the tick-protective antigens that have been identified for the formulation of anti-tick vaccines along with the effects of these vaccines on the control of tick-borne diseases.
Collapse
Affiliation(s)
- Biswajit Bhowmick
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Qian Han
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, China
| |
Collapse
|
30
|
Wen S, Wang F, Ji Z, Pan Y, Jian M, Bi Y, Zhou G, Luo L, Chen T, Li L, Ding Z, Abi ME, Liu A, Bao F. Salp15, a Multifunctional Protein From Tick Saliva With Potential Pharmaceutical Effects. Front Immunol 2020; 10:3067. [PMID: 31998324 PMCID: PMC6968165 DOI: 10.3389/fimmu.2019.03067] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/16/2019] [Indexed: 12/30/2022] Open
Abstract
Ixodes ticks are the main vectors for a number of zoonotic diseases, including Lyme disease. Ticks secrete saliva directly into a mammalian host while feeding on the host's blood. This action serves to modulate host immunity and coagulation, thus allowing ticks to attach and feed upon their host. One of the most extensively studied components of tick saliva is Salp15. Research has shown that this protein binds specifically to CD4 molecules on the surface of T lymphocytes, interferes with TCR-mediated signaling transduction, inhibits CD4+ T cell activation and proliferation, and impedes the secretion of interleukin 2 (IL-2). Salp15 also binds specifically to dendritic cell dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) to up-regulate the expression of CD73 in regulatory T cells. Collectively, these findings render this salivary protein a potential candidate for a range of therapeutic applications. Here, we discuss our current understanding of Salp15 and the mechanisms that might be used to treat disease.
Collapse
Affiliation(s)
- Shiyuan Wen
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China.,The Center of Tropical Diseases, The Institute for Tropical Medicine, Kunming Medical University, Kunming, China.,Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases, Kunming, China.,The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Feng Wang
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China.,The Center of Tropical Diseases, The Institute for Tropical Medicine, Kunming Medical University, Kunming, China.,Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases, Kunming, China
| | - Zhenhua Ji
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - YingYi Pan
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Miaomiao Jian
- The Center of Tropical Diseases, The Institute for Tropical Medicine, Kunming Medical University, Kunming, China.,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - YunFeng Bi
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China.,The Center of Tropical Diseases, The Institute for Tropical Medicine, Kunming Medical University, Kunming, China.,Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases, Kunming, China
| | - Guozhong Zhou
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China.,The Center of Tropical Diseases, The Institute for Tropical Medicine, Kunming Medical University, Kunming, China.,Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases, Kunming, China
| | - Lisha Luo
- The Center of Tropical Diseases, The Institute for Tropical Medicine, Kunming Medical University, Kunming, China.,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Taigui Chen
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Lianbao Li
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Zhe Ding
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Manzama-Esso Abi
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China
| | - Aihua Liu
- The Center of Tropical Diseases, The Institute for Tropical Medicine, Kunming Medical University, Kunming, China.,Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases, Kunming, China.,Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, China
| | - Fukai Bao
- Department of Microbiology and Immunology, Kunming Medical University, Kunming, China.,The Center of Tropical Diseases, The Institute for Tropical Medicine, Kunming Medical University, Kunming, China.,Yunnan Demonstration Base of International Science and Technology Cooperation for Tropical Diseases, Kunming, China
| |
Collapse
|
31
|
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
|
32
|
Sanchez-Vicente S, Tagliafierro T, Coleman JL, Benach JL, Tokarz R. Polymicrobial Nature of Tick-Borne Diseases. mBio 2019; 10:e02055-19. [PMID: 31506314 PMCID: PMC6737246 DOI: 10.1128/mbio.02055-19] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 01/08/2023] Open
Abstract
Tick-borne diseases have doubled in the last 12 years, and their geographic distribution has spread as well. The clinical spectrum of tick-borne diseases can range from asymptomatic to fatal infections, with a disproportionate incidence in children and the elderly. In the last few years, new agents have been discovered, and genetic changes have helped in the spread of pathogens and ticks. Polymicrobial infections, mostly in Ixodes scapularis, can complicate diagnostics and augment disease severity. Amblyomma americanum ticks have expanded their range, resulting in a dynamic and complex situation, possibly fueled by climate change. To document these changes, using molecular biology strategies for pathogen detection, an assessment of 12 microbes (9 pathogens and 3 symbionts) in three species of ticks was done in Suffolk County, New York. At least one agent was detected in 63% of I. scapularis ticksBorrelia burgdorferi was the most prevalent pathogen (57% in adults; 27% in nymphs), followed by Babesia microti (14% in adults; 15% in nymphs), Anaplasma phagocytophilum (14% in adults; 2% in nymphs), Borrelia miyamotoi (3% in adults), and Powassan virus (2% in adults). Polymicrobial infections were detected in 22% of I. scapularis ticks, with coinfections of B. burgdorferi and B. microti (9%) and of B. burgdorferi and A. phagocytophilum (7%). Three Ehrlichia species were detected in 4% of A. americanum ticks. The rickettsiae constituted the largest prokaryotic biomass of all the ticks tested and included Rickettsia amblyommatis, Rickettsia buchneri, and Rickettsia montanensis The high rates of polymicrobial infection in ticks present an opportunity to study the biological interrelationships of pathogens and their vectors.IMPORTANCE Tick-borne diseases have increased in prevalence in the United States and abroad. The reasons for these increases are multifactorial, but climate change is likely to be a major factor. One of the main features of the increase is the geographic expansion of tick vectors, notably Amblyomma americanum, which has brought new pathogens to new areas. The clinical spectrum of tick-borne diseases can range from asymptomatic to fatal infections, with a disproportionate incidence in children and the elderly. In addition, new pathogens that are cotransmitted by Ixodes scapularis have been discovered and have led to difficult diagnoses and to disease severity. Of these, Borrelia burgdorferi, the agent of Lyme disease, continues to be the most frequently transmitted pathogen. However, Babesia microti, Borrelia miyamotoi (another spirochete), Anaplasma phagocytophilum, and Powassan virus are frequent cotransmitted agents. Polymicrobial infection has important consequences for the diagnosis and management of tick-borne diseases.
Collapse
Affiliation(s)
- Santiago Sanchez-Vicente
- Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Teresa Tagliafierro
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - James L Coleman
- Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Jorge L Benach
- Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Rafal Tokarz
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, New York, USA
| |
Collapse
|
33
|
Rego ROM, Trentelman JJA, Anguita J, Nijhof AM, Sprong H, Klempa B, Hajdusek O, Tomás-Cortázar J, Azagi T, Strnad M, Knorr S, Sima R, Jalovecka M, Fumačová Havlíková S, Ličková M, Sláviková M, Kopacek P, Grubhoffer L, Hovius JW. Counterattacking the tick bite: towards a rational design of anti-tick vaccines targeting pathogen transmission. Parasit Vectors 2019; 12:229. [PMID: 31088506 PMCID: PMC6518728 DOI: 10.1186/s13071-019-3468-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/29/2019] [Indexed: 02/07/2023] Open
Abstract
Hematophagous arthropods are responsible for the transmission of a variety of pathogens that cause disease in humans and animals. Ticks of the Ixodes ricinus complex are vectors for some of the most frequently occurring human tick-borne diseases, particularly Lyme borreliosis and tick-borne encephalitis virus (TBEV). The search for vaccines against these diseases is ongoing. Efforts during the last few decades have primarily focused on understanding the biology of the transmitted viruses, bacteria and protozoans, with the goal of identifying targets for intervention. Successful vaccines have been developed against TBEV and Lyme borreliosis, although the latter is no longer available for humans. More recently, the focus of intervention has shifted back to where it was initially being studied which is the vector. State of the art technologies are being used for the identification of potential vaccine candidates for anti-tick vaccines that could be used either in humans or animals. The study of the interrelationship between ticks and the pathogens they transmit, including mechanisms of acquisition, persistence and transmission have come to the fore, as this knowledge may lead to the identification of critical elements of the pathogens' life-cycle that could be targeted by vaccines. Here, we review the status of our current knowledge on the triangular relationships between ticks, the pathogens they carry and the mammalian hosts, as well as methods that are being used to identify anti-tick vaccine candidates that can prevent the transmission of tick-borne pathogens.
Collapse
Affiliation(s)
- Ryan O. M. Rego
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
| | - Jos J. A. Trentelman
- Amsterdam UMC, Location AMC, Center for Experimental and Molecular Medicine, Amsterdam, The Netherlands
| | - Juan Anguita
- CIC bioGUNE, 48160 Derio, Spain
- Ikerbasque, Basque Foundation for Science, 48012 Bilbao, Spain
| | - Ard M. Nijhof
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Hein Sprong
- Centre for Zoonoses and Environmental Microbiology, Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Boris Klempa
- Institute of Virology, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Ondrej Hajdusek
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
| | | | - Tal Azagi
- Centre for Zoonoses and Environmental Microbiology, Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Martin Strnad
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
- Faculty of Science, University of South Bohemia, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
| | - Sarah Knorr
- Institute for Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Radek Sima
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
| | - Marie Jalovecka
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
- Faculty of Science, University of South Bohemia, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
| | - Sabína Fumačová Havlíková
- Institute of Virology, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Martina Ličková
- Institute of Virology, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Monika Sláviková
- Institute of Virology, Biomedical Research Center of the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Petr Kopacek
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
- Faculty of Science, University of South Bohemia, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
| | - Libor Grubhoffer
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
- Faculty of Science, University of South Bohemia, Branišovská 31, 37005 Ceske Budejovice, Czech Republic
| | - Joppe W. Hovius
- Amsterdam UMC, Location AMC, Center for Experimental and Molecular Medicine, Amsterdam, The Netherlands
| |
Collapse
|
34
|
Turck JW, Taank V, Neelakanta G, Sultana H. Ixodes scapularis Src tyrosine kinase facilitates Anaplasma phagocytophilum survival in its arthropod vector. Ticks Tick Borne Dis 2019; 10:838-847. [PMID: 31000483 DOI: 10.1016/j.ttbdis.2019.04.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 03/19/2019] [Accepted: 04/03/2019] [Indexed: 11/25/2022]
Abstract
Anaplasma phagocytophilum, the agent of human anaplasmosis, is an obligate intracellular bacterium that uses multiple survival strategies to persist in Ixodes scapularis ticks. Our previous study showed that A. phagocytophilum efficiently induced the tyrosine phosphorylation of several Ixodes proteins that includes extended phosphorylation of actin at tyrosine residue Y178. In order to identify the tyrosine kinase responsible for the A. phagocytophilum induced tyrosine phosphorylation of proteins, we combed the I. scapularis genome and identified a non-receptor Src tyrosine kinase ortholog. I. scapularis Src kinase showed high degree of amino acid sequence conservation with Dsrc from Drosophila melanogaster. We noted that at different developmental stages of I. scapularis ticks, larvae expressed significantly higher levels of src transcripts in comparison to the other stages. We found that A. phagocytophilum significantly reduced Src levels in unfed nymphs and in nymphs while blood feeding (48 h during feeding) in comparison to the levels noted to relative uninfected controls. However, A. phagocytophilum increased Src levels in fully engorged larvae and nymphs (48 h post feeding) and in vitro tick cells in comparison to the relative uninfected controls. Inhibition of Src kinase expression and activity by treatment with src-dsRNA or Src-inhibitor, respectively, significantly reduced A. phagocytophilum loads in ticks and tick cells. Overall, our study provides evidence for the important role of I. scapularis Src kinase in facilitating A. phagocytophilum colonization and survival in the arthropod vector.
Collapse
Affiliation(s)
- Jeremy W Turck
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - Vikas Taank
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA; Center for Molecular Medicine, College of Sciences, Old Dominion University, Norfolk, VA, USA.
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA; Center for Molecular Medicine, College of Sciences, Old Dominion University, Norfolk, VA, USA.
| |
Collapse
|
35
|
Cabezas-Cruz A, Espinosa P, Alberdi P, de la Fuente J. Tick-Pathogen Interactions: The Metabolic Perspective. Trends Parasitol 2019; 35:316-328. [PMID: 30711437 DOI: 10.1016/j.pt.2019.01.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/08/2019] [Accepted: 01/09/2019] [Indexed: 01/03/2023]
Abstract
The first tick genome published in 2016 provided an invaluable tool for studying the molecular basis of tick-pathogen interactions. Metabolism is a key element in host-pathogen interactions. However, our knowledge of tick-pathogen metabolic interactions is very limited. Recently, a systems biology approach, using omics datasets, has revealed that tick-borne pathogen infection induces transcriptional reprograming affecting several metabolic pathways in ticks, facilitating infection, multiplication, and transmission. Results suggest that the response of tick cells to tick-borne pathogens is associated with tolerance to infection. Here we review our current understanding of the modulation of tick metabolism by tick-borne pathogens, with a focus on the model intracellular bacterium Anaplasma phagocytophilum.
Collapse
Affiliation(s)
- Alejandro Cabezas-Cruz
- UMR BIPAR, INRA, Ecole Nationale Vétérinaire d'Alfort, ANSES, Université Paris-Est, Maisons-Alfort, France.
| | - Pedro Espinosa
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain
| | - Pilar Alberdi
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain
| | - 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.
| |
Collapse
|
36
|
Taank V, Zhou W, Zhuang X, Anderson JF, Pal U, Sultana H, Neelakanta G. Characterization of tick organic anion transporting polypeptides (OATPs) upon bacterial and viral infections. Parasit Vectors 2018; 11:593. [PMID: 30428915 PMCID: PMC6236954 DOI: 10.1186/s13071-018-3160-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/22/2018] [Indexed: 12/21/2022] Open
Abstract
Background Ixodes scapularis organic anion transporting polypeptides (OATPs) play important roles in tick-rickettsial pathogen interactions. In this report, we characterized the role of these conserved molecules in ticks infected with either Lyme disease agent Borrelia burgdorferi or tick-borne Langat virus (LGTV), a pathogen closely related to tick-borne encephalitis virus (TBEV). Results Quantitative real-time polymerase chain reaction analysis revealed no significant changes in oatps gene expression upon infection with B. burgdorferi in unfed ticks. Synchronous infection of unfed nymphal ticks with LGTV in vitro revealed no significant changes in oatps gene expression. However, expression of specific oatps was significantly downregulated upon LGTV infection of tick cells in vitro. Treatment of tick cells with OATP inhibitor significantly reduced LGTV loads, kynurenine amino transferase (kat), a gene involved in the production of tryptophan metabolite xanthurenic acid (XA), levels and expression of several oatps in tick cells. Furthermore, bioinformatics characterization of OATPs from some of the medically important vectors including ticks, mosquitoes and lice revealed the presence of several glycosylation, phosphorylation and myristoylation sites. Conclusions This study provides additional evidence on the role of arthropod OATPs in vector-intracellular pathogen interactions. Electronic supplementary material The online version of this article (10.1186/s13071-018-3160-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vikas Taank
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - Wenshuo Zhou
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - Xuran Zhuang
- Department of Veterinary Medicine, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - John F Anderson
- Department of Entomology, Connecticut Agricultural Experiment Station, New Haven, CT, USA
| | - Utpal Pal
- Department of Veterinary Medicine, Virginia-Maryland Regional College of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA.,Center for Molecular Medicine, College of Sciences, Old Dominion University, Norfolk, VA, USA
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA. .,Center for Molecular Medicine, College of Sciences, Old Dominion University, Norfolk, VA, USA.
| |
Collapse
|
37
|
Kim D, Jaworski DC, Cheng C, Nair AD, Ganta RR, Herndon N, Brown S, Park Y. The transcriptome of the lone star tick, Amblyomma americanum, reveals molecular changes in response to infection with the pathogen, Ehrlichia chaffeensis. JOURNAL OF ASIA-PACIFIC ENTOMOLOGY 2018; 21:852-863. [PMID: 34316264 PMCID: PMC8312692 DOI: 10.1016/j.aspen.2018.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The lone star tick, Amblyomma americanum, is an obligatory ectoparasite of many vertebrates and the primary vector of Ehrlichia chaffeensis, the causative agent of human monocytic ehrlichiosis. This study aimed to investigate the comparative transcriptomes of A. americanum underlying the processes of pathogen acquisition and of immunity towards the pathogen. Differential expression of the whole body transcripts in six different treatments were compared: females and males that were E. chaffeensis non-exposed, E. chaffeensis-exposed/uninfected, and E. chaffeensis-exposed/infected. The Trinity assembly pipeline produced 140,574 transcripts from trimmed and filtered total raw sequence reads (approximately 117M reads). The gold transcript set of the transcriptome data was established to minimize noise by retaining only transcripts homologous to official peptide sets of Ixodes scapularis and A. americanum ESTs and transcripts covered with high enough frequency from the raw data. Comparison of the gene ontology term enrichment analyses for the six groups tested here revealed an up-regulation of genes for defense responses against the pathogen and for the supply of intracellular Ca++ for pathogen proliferation in the pathogen-exposed ticks. Analyses of differential expression, focused on functional subcategories including immune, sialome, neuropeptides, and G protein-coupled receptor, revealed that E. chaffeensis-exposed ticks exhibited an upregulation of transcripts involved in the immune deficiency (IMD) pathway, antimicrobial peptides, Kunitz, an insulin-like peptide, and bursicon receptor over unexposed ones, while transcripts for metalloprotease were down-regulated in general. This study found that ticks exhibit enhanced expression of genes responsible for defense against E. chaffeensis.
Collapse
Affiliation(s)
- Donghun Kim
- Department of Entomology, Kansas State University, Manhattan, KS 66506, USA
| | - Deborah C. Jaworski
- Center of Excellence for Vector-Borne Diseases, Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, USA
| | - Chuanmin Cheng
- Center of Excellence for Vector-Borne Diseases, Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, USA
| | - Arathy D.S. Nair
- Center of Excellence for Vector-Borne Diseases, Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, USA
| | - Roman R. Ganta
- Center of Excellence for Vector-Borne Diseases, Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506, USA
| | - Nic Herndon
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Susan Brown
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Yoonseong Park
- Department of Entomology, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
38
|
Estrada-Peña A, Villar M, Artigas-Jerónimo S, López V, Alberdi P, Cabezas-Cruz A, de la Fuente J. Use of Graph Theory to Characterize Human and Arthropod Vector Cell Protein Response to Infection With Anaplasma phagocytophilum. Front Cell Infect Microbiol 2018; 8:265. [PMID: 30123779 PMCID: PMC6086010 DOI: 10.3389/fcimb.2018.00265] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/13/2018] [Indexed: 12/30/2022] Open
Abstract
One of the major challenges in modern biology is the use of large omics datasets for the characterization of complex processes such as cell response to infection. These challenges are even bigger when analyses need to be performed for comparison of different species including model and non-model organisms. To address these challenges, the graph theory was applied to characterize the tick vector and human cell protein response to infection with Anaplasma phagocytophilum, the causative agent of human granulocytic anaplasmosis. A network of interacting proteins and cell processes clustered in biological pathways, and ranked with indexes representing the topology of the proteome was prepared. The results demonstrated that networks of functionally interacting proteins represented in both infected and uninfected cells can describe the complete set of host cell processes and metabolic pathways, providing a deeper view of the comparative host cell response to pathogen infection. The results demonstrated that changes in the tick proteome were driven by modifications in protein representation in response to A. phagocytophilum infection. Pathogen infection had a higher impact on tick than human proteome. Since most proteins were linked to several cell processes, the changes in protein representation affected simultaneously different biological pathways. The method allowed discerning cell processes that were affected by pathogen infection from those that remained unaffected. The results supported that human neutrophils but not tick cells limit pathogen infection through differential representation of ras-related proteins. This methodological approach could be applied to other host-pathogen models to identify host derived key proteins in response to infection that may be used to develop novel control strategies for arthropod-borne pathogens.
Collapse
Affiliation(s)
| | - Margarita Villar
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC), CSIC, Universidad de Castilla-La Mancha (UCLM), Junta de Comunidades de Castilla - La Mancha (JCCM), Ciudad Real, Spain
| | - Sara Artigas-Jerónimo
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC), CSIC, Universidad de Castilla-La Mancha (UCLM), Junta de Comunidades de Castilla - La Mancha (JCCM), Ciudad Real, Spain
| | - Vladimir López
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC), CSIC, Universidad de Castilla-La Mancha (UCLM), Junta de Comunidades de Castilla - La Mancha (JCCM), Ciudad Real, Spain
| | - Pilar Alberdi
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC), CSIC, Universidad de Castilla-La Mancha (UCLM), Junta de Comunidades de Castilla - La Mancha (JCCM), Ciudad Real, Spain
| | - Alejandro Cabezas-Cruz
- UMR Biologie Moléculaire et Immunologie Parasitaires (BIPAR), INRA, Agence Nationale de Sécurité Sanitairede l'Alimentation, de l'Environnement et du Travail (ANSES), Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France.,Faculty of Science, University of South Bohemia, Ceské Budějovice, Czechia.,Institute of Parasitology, Biology Center, Czech Academy of Sciences, Ceské Budějovice, Czechia
| | - José de la Fuente
- Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain.,Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, United States
| |
Collapse
|
39
|
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
|
40
|
Arthropod EVs mediate dengue virus transmission through interaction with a tetraspanin domain containing glycoprotein Tsp29Fb. Proc Natl Acad Sci U S A 2018; 115:E6604-E6613. [PMID: 29946031 DOI: 10.1073/pnas.1720125115] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Dengue virus (DENV) is a mosquito-borne flavivirus that causes dengue fever in humans, worldwide. Using in vitro cell lines derived from Aedes albopictus and Aedes aegypti, the primary vectors of DENV, we report that DENV2/DENV3-infected cells secrete extracellular vesicles (EVs), including exosomes, containing infectious viral RNA and proteins. A full-length DENV2 genome, detected in arthropod EVs, was infectious to naïve mosquito and mammalian cells, including human-skin keratinocytes and blood endothelial cells. Cryo-electron microscopy showed mosquito EVs with a size range from 30 to 250 nm. Treatments with RNase A, Triton X-100, and 4G2 antibody-bead binding assays showed that infectious DENV2-RNA and proteins are contained inside EVs. Viral plaque formation and dilution assays also showed securely contained infectious viral RNA and proteins in EVs are transmitted to human cells. Up-regulated HSP70 upon DENV2 infection showed no role in viral replication and transmission through EVs. In addition, qRT-PCR and immunoblotting results revealed that DENV2 up-regulates expression of a mosquito tetraspanin-domain-containing glycoprotein, designated as Tsp29Fb, in A. aegypti mosquitoes, cells, and EVs. RNAi-mediated silencing and antibody blocking of Tsp29Fb resulted in reduced DENV2 loads in both mosquito cells and EVs. Immunoprecipitation showed Tsp29Fb to directly interact with DENV2 E-protein. Furthermore, treatment with GW4869 (exosome-release inhibitor) affected viral burden, direct interaction of Tsp29Fb with E-protein and EV-mediated transmission of viral RNA and proteins to naïve human cells. In summary, we report a very important finding on EV-mediated transmission of DENV2 from arthropod to mammalian cells through interactions with an arthropod EVs-enriched marker Tsp29Fb.
Collapse
|
41
|
Neelakanta G, Sultana H, Sonenshine DE, Andersen JF. Identification and characterization of a histamine-binding lipocalin-like molecule from the relapsing fever tick Ornithodoros turicata. INSECT MOLECULAR BIOLOGY 2018; 27:177-187. [PMID: 29164729 DOI: 10.1111/imb.12362] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Lipocalins are low molecular weight membrane transporters that are abundantly expressed in the salivary glands and other tissues of ticks. In this study, we identified a lipocalin-like molecule, designated as otlip, from the soft ticks Ornithodoros turicata, the vector for the relapsing fever causing spirochete Borrelia turicatae. We noted that the expression of otlip was developmentally regulated, with adult ticks expressing significantly higher levels in comparison to the larvae or nymphal ticks. Expression of otlip was evident in both fed and unfed O. turicata ticks, with significantly increased expression in the salivary glands in comparison to the midgut or ovary tissues. High conservation of the biogenic amine-binding motif was evident in the deduced primary amino acid sequence of Otlip. Protein modelling of Otlip revealed conservation of most of the residues involved in binding histamine or serotonin ligand. In vitro assays demonstrated binding of recombinant Otlip with histamine. Furthermore, prediction of post-translational modifications revealed that Otlip contained phosphorylation and myristoylation sites. Taken together, our study not only provides evidence for the presence of a lipocalin-like molecule in O. turicata ticks but also suggests a role for this molecule in the salivary glands of this medically important vector.
Collapse
Affiliation(s)
- G Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
- Center for Molecular Medicine, Old Dominion University, Norfolk, VA, USA
| | - H Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
- Center for Molecular Medicine, Old Dominion University, Norfolk, VA, USA
| | - D E Sonenshine
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
- The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, MA, USA
| | - J F Andersen
- The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, MA, USA
| |
Collapse
|
42
|
Zhou W, Woodson M, Neupane B, Bai F, Sherman MB, Choi KH, Neelakanta G, Sultana H. Exosomes serve as novel modes of tick-borne flavivirus transmission from arthropod to human cells and facilitates dissemination of viral RNA and proteins to the vertebrate neuronal cells. PLoS Pathog 2018; 14:e1006764. [PMID: 29300779 PMCID: PMC5754134 DOI: 10.1371/journal.ppat.1006764] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 11/20/2017] [Indexed: 11/18/2022] Open
Abstract
Molecular determinants and mechanisms of arthropod-borne flavivirus transmission to the vertebrate host are poorly understood. In this study, we show for the first time that a cell line from medically important arthropods, such as ticks, secretes extracellular vesicles (EVs) including exosomes that mediate transmission of flavivirus RNA and proteins to the human cells. Our study shows that tick-borne Langat virus (LGTV), a model pathogen closely related to tick-borne encephalitis virus (TBEV), profusely uses arthropod exosomes for transmission of viral RNA and proteins to the human- skin keratinocytes and blood endothelial cells. Cryo-electron microscopy showed the presence of purified arthropod/neuronal exosomes with the size range of 30 to 200 nm in diameter. Both positive and negative strands of LGTV RNA and viral envelope-protein were detected inside exosomes derived from arthropod, murine and human cells. Detection of Nonstructural 1 (NS1) protein in arthropod and neuronal exosomes further suggested that exosomes contain viral proteins. Viral RNA and proteins in exosomes derived from tick and mammalian cells were secured, highly infectious and replicative in all tested evaluations. Treatment with GW4869, a selective inhibitor that blocks exosome release affected LGTV loads in both arthropod and mammalian cell-derived exosomes. Transwell-migration assays showed that exosomes derived from infected-brain-microvascular endothelial cells (that constitute the blood-brain barrier) facilitated LGTV RNA and protein transmission, crossing of the barriers and infection of neuronal cells. Neuronal infection showed abundant loads of both tick-borne LGTV and mosquito-borne West Nile virus RNA in exosomes. Our data also suggest that exosome-mediated LGTV viral transmission is clathrin-dependent. Collectively, our results suggest that flaviviruses uses arthropod-derived exosomes as a novel means for viral RNA and protein transmission from the vector, and the vertebrate exosomes for dissemination within the host that may subsequently allow neuroinvasion and neuropathogenesis.
Collapse
MESH Headings
- Animals
- Arthropod Vectors/cytology
- Arthropod Vectors/ultrastructure
- Arthropod Vectors/virology
- Cell Line
- Cells, Cultured
- Cerebral Cortex/cytology
- Cerebral Cortex/pathology
- Cerebral Cortex/ultrastructure
- Cerebral Cortex/virology
- Chlorocebus aethiops
- Coculture Techniques
- Cryoelectron Microscopy
- Embryo, Mammalian/cytology
- Encephalitis Viruses, Tick-Borne/pathogenicity
- Encephalitis Viruses, Tick-Borne/physiology
- Encephalitis Viruses, Tick-Borne/ultrastructure
- Encephalitis, Tick-Borne/pathology
- Encephalitis, Tick-Borne/transmission
- Encephalitis, Tick-Borne/virology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/pathology
- Endothelium, Vascular/ultrastructure
- Endothelium, Vascular/virology
- Exosomes/ultrastructure
- Exosomes/virology
- Host-Parasite Interactions
- Host-Pathogen Interactions
- Humans
- Ixodes/cytology
- Ixodes/ultrastructure
- Ixodes/virology
- Keratinocytes/cytology
- Keratinocytes/pathology
- Keratinocytes/ultrastructure
- Keratinocytes/virology
- Mice
- Mice, Inbred C57BL
- Models, Biological
- Neurons/cytology
- Neurons/pathology
- Neurons/ultrastructure
- Neurons/virology
- RNA, Viral/metabolism
- Viral Proteins/metabolism
Collapse
Affiliation(s)
- Wenshuo Zhou
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States of America
| | - Michael Woodson
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Biswas Neupane
- Department of Biological Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States of America
| | - Fengwei Bai
- Department of Biological Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States of America
| | - Michael B. Sherman
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States of America
- Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Kyung H. Choi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States of America
- Center for Molecular Medicine, Old Dominion University, Norfolk, VA, United States of America
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States of America
- Center for Molecular Medicine, Old Dominion University, Norfolk, VA, United States of America
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, VA, United States of America
- * E-mail:
| |
Collapse
|
43
|
Taank V, Dutta S, Dasgupta A, Steeves TK, Fish D, Anderson JF, Sultana H, Neelakanta G. Human rickettsial pathogen modulates arthropod organic anion transporting polypeptide and tryptophan pathway for its survival in ticks. Sci Rep 2017; 7:13256. [PMID: 29038575 PMCID: PMC5643405 DOI: 10.1038/s41598-017-13559-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/25/2017] [Indexed: 12/19/2022] Open
Abstract
The black-legged tick Ixodes scapularis transmits the human anaplasmosis agent, Anaplasma phagocytophilum. In this study, we show that A. phagocytophilum specifically up-regulates I. scapularis organic anion transporting polypeptide, isoatp4056 and kynurenine amino transferase (kat), a gene involved in the production of tryptophan metabolite xanthurenic acid (XA), for its survival in ticks. RNAi analysis revealed that knockdown of isoatp4056 expression had no effect on A. phagocytophilum acquisition from the murine host but affected the bacterial survival in tick cells. Knockdown of the expression of kat mRNA alone or in combination with isoatp4056 mRNA significantly affected A. phagocytophilum survival and isoatp4056 expression in tick cells. Exogenous addition of XA induces isoatp4056 expression and A. phagocytophilum burden in both tick salivary glands and tick cells. Electrophoretic mobility shift assays provide further evidence that A. phagocytophilum and XA influences isoatp4056 expression. Collectively, this study provides important novel information in understanding the interplay between molecular pathways manipulated by a rickettsial pathogen to survive in its arthropod vector.
Collapse
Affiliation(s)
- Vikas Taank
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - Shovan Dutta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - Amrita Dasgupta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA.,Skin of Color Research Institute, Hampton University, Hampton, VA, USA
| | - Tanner K Steeves
- School of Public Health, Yale University School of Medicine, New Haven, CT, USA
| | - Durland Fish
- School of Public Health, Yale University School of Medicine, New Haven, CT, 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
|
44
|
Khanal S, Sultana H, Catravas JD, Carlyon JA, Neelakanta G. Anaplasma phagocytophilum infection modulates expression of megakaryocyte cell cycle genes through phosphatidylinositol-3-kinase signaling. PLoS One 2017; 12:e0182898. [PMID: 28797056 PMCID: PMC5552339 DOI: 10.1371/journal.pone.0182898] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/20/2017] [Indexed: 12/19/2022] Open
Abstract
Anaplasma phagocytophilum, the agent of human granulocytic anaplasmosis infects neutrophils and other cells from hematopoietic origin. Using human megakaryocytic cell line, MEG-01, we show that expression of cell cycle genes in these cells are altered upon A. phagocytophilum infection. Expression of several cell cycle genes in MEG-01 cells was significantly up regulated at early and then down regulated at later stages of A. phagocytophilum infection. Lactate dehydrogenase (LDH) assays revealed reduced cellular cytotoxicity in MEG-01 cells upon A. phagocytophilum infection. The levels of both PI3KCA (p110 alpha, catalytic subunit) and PI3KR1 (p85, regulatory subunit) of Class I PI3 kinases and phosphorylated protein kinase B (Akt/PKB) and inhibitory kappa B (IκB) were elevated at both early and late stages of A. phagocytophilum infection. Inhibition of PI3 kinases with LY294002 treatment resulted in significant reduction in the expression of tested cell cycle genes, A. phagocytophilum burden and phosphorylated Akt levels in these MEG-01 cells. Collectively, these results suggest a role for PI3K-Akt-NF-κB signaling pathway in the modulation of megakaryocyte cell cycle genes upon A. phagocytophilum infection.
Collapse
Affiliation(s)
- Supreet Khanal
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States of America
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States of America
- Center for Molecular Medicine, College of Sciences, Old Dominion University, Norfolk, VA, United States of America
| | - John D. Catravas
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, United States of America
- School of Medical Diagnostic and Translational Sciences, College of Health Sciences, Old Dominion University, Norfolk, VA, United States of America
| | - Jason A. Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, United States of America
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, United States of America
- Center for Molecular Medicine, College of Sciences, Old Dominion University, Norfolk, VA, United States of America
- * E-mail:
| |
Collapse
|
45
|
Actin Tyrosine-53-Phosphorylation in Neuronal Maturation and Synaptic Plasticity. J Neurosci 2017; 36:5299-313. [PMID: 27170127 DOI: 10.1523/jneurosci.2649-15.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 03/31/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Rapid reorganization and stabilization of the actin cytoskeleton in dendritic spines enables cellular processes underlying learning, such as long-term potentiation (LTP). Dendritic spines are enriched in exceptionally short and dynamic actin filaments, but the studies so far have not revealed the molecular mechanisms underlying the high actin dynamics in dendritic spines. Here, we show that actin in dendritic spines is dynamically phosphorylated at tyrosine-53 (Y53) in rat hippocampal and cortical neurons. Our findings show that actin phosphorylation increases the turnover rate of actin filaments and promotes the short-term dynamics of dendritic spines. During neuronal maturation, actin phosphorylation peaks at the first weeks of morphogenesis, when dendritic spines form, and the amount of Y53-phosphorylated actin decreases when spines mature and stabilize. Induction of LTP transiently increases the amount of phosphorylated actin and LTP induction is deficient in neurons expressing mutant actin that mimics phosphorylation. Actin phosphorylation provides a molecular mechanism to maintain the high actin dynamics in dendritic spines during neuronal development and to induce fast reorganization of the actin cytoskeleton in synaptic plasticity. In turn, dephosphorylation of actin is required for the stabilization of actin filaments that is necessary for proper dendritic spine maturation and LTP maintenance. SIGNIFICANCE STATEMENT Dendritic spines are small protrusions from neuronal dendrites where the postsynaptic components of most excitatory synapses reside. Precise control of dendritic spine morphology and density is critical for normal brain function. Accordingly, aberrant spine morphology is linked to many neurological diseases. The actin cytoskeleton is a structural element underlying the proper morphology of dendritic spines. Therefore, defects in the regulation of the actin cytoskeleton in neurons have been implicated in neurological diseases. Here, we revealed a novel mechanism for regulating neuronal actin cytoskeleton that explains the specific organization and dynamics of actin in spines. The better we understand the regulation of the dendritic spine morphology, the better we understand what goes wrong in neurological diseases.
Collapse
|
46
|
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
|
47
|
Vora A, Taank V, Dutta SM, Anderson JF, Fish D, Sonenshine DE, Catravas JD, Sultana H, Neelakanta G. Ticks elicit variable fibrinogenolytic activities upon feeding on hosts with different immune backgrounds. Sci Rep 2017; 7:44593. [PMID: 28300174 PMCID: PMC5353578 DOI: 10.1038/srep44593] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/10/2017] [Indexed: 12/11/2022] Open
Abstract
Ticks secrete several anti-hemostatic factors in their saliva to suppress the host innate and acquired immune defenses against infestations. Using Ixodes scapularis ticks and age-matched mice purchased from two independent commercial vendors with two different immune backgrounds as a model, we show that ticks fed on immunodeficient animals demonstrate decreased fibrinogenolytic activity in comparison to ticks fed on immunocompetent animals. Reduced levels of D-dimer (fibrin degradation product) were evident in ticks fed on immunodeficient animals in comparison to ticks fed on immunocompetent animals. Increased engorgement weights were noted for ticks fed on immunodeficient animals in comparison to ticks fed on immunocompetent animals. Furthermore, the LC-MS/MS and quantitative real-time-PCR analysis followed by inhibitor and antibody-blocking assays revealed that the arthropod HSP70-like molecule contributes to differential fibrinogenolysis during tick feeding. Collectively, these results not only indicate that ticks elicit variable fibrinogenolysis upon feeding on hosts with different immune backgrounds but also provide insights for the novel role of arthropod HSP70-like molecule in fibrinogenolysis during blood feeding.
Collapse
Affiliation(s)
- Ashish Vora
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - Vikas Taank
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - Sucharita M Dutta
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, USA
| | - John F Anderson
- Department of Entomology, Connecticut Agricultural Experiment Station, New Haven, CT, USA
| | - Durland Fish
- School of Public Health, Yale University School of Medicine, New Haven, CT, USA
| | - Daniel E Sonenshine
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - John D Catravas
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, VA, USA.,School of Medical Diagnostic and Translational Sciences, College of Health Sciences, Old Dominion University, Norfolk, VA, USA
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA.,Center for Molecular Medicine, College of Sciences, Old Dominion University, Norfolk, VA, USA
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA.,Center for Molecular Medicine, College of Sciences, Old Dominion University, Norfolk, VA, USA
| |
Collapse
|
48
|
Cabezas-Cruz A, Estrada-Peña A, Rego ROM, De la Fuente J. Tick-Pathogen Ensembles: Do Molecular Interactions Lead Ecological Innovation? Front Cell Infect Microbiol 2017; 7:74. [PMID: 28348982 PMCID: PMC5346557 DOI: 10.3389/fcimb.2017.00074] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 02/27/2017] [Indexed: 11/18/2022] Open
Affiliation(s)
- Alejandro Cabezas-Cruz
- UMR BIPAR, Animal Health Laboratory, ANSES, Institut National de la Recherche Agronomique, ENVAMaisons Alfort, France; Department of Parasitology, Faculty of Science, University of South BohemiaČeské Budějovice, Czechia; Biology Center, Institute of Parasitology, Czech Academy of SciencesČeské Budějovice, Czechia
| | | | - Ryan O M Rego
- Department of Parasitology, Faculty of Science, University of South BohemiaČeské Budějovice, Czechia; Biology Center, Institute of Parasitology, Czech Academy of SciencesČeské Budějovice, Czechia
| | - 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 UniversityStillwater, OK, USA
| |
Collapse
|
49
|
Neelakanta G, Sultana H, Sonenshine DE, Marconi RT. An In Vitro Blood-Feeding Method Revealed Differential Borrelia turicatae (Spirochaetales: Spirochaetaceae) Gene Expression After Spirochete Acquisition and Colonization in the Soft Tick Ornithodoros turicata (Acari: Argasidae). JOURNAL OF MEDICAL ENTOMOLOGY 2017; 54:441-449. [PMID: 28399292 DOI: 10.1093/jme/tjw171] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/20/2016] [Indexed: 06/07/2023]
Abstract
In the Midwestern, Southwestern, and Southern part of the United States, the soft tick Ornithodoros turicata transmits the spirochete Borrelia turicatae, the causative agent of relapsing fever in humans. In this study, we report a simplified and an efficient method of in vitro feeding to evaluate O. turicata-B. turicatae interactions. Both nymphal and adult female ticks successfully acquired spirochetes upon in vitro feeding on the B. turicatae-infected blood. We also noted transstadial transmission of spirochetes to adult ticks that were molted from nymphs fed on B. turicatae-infected blood. A differential expression pattern for some of the B. turicatae genes was evident after acquisition and colonization of the vector. The levels of arthropod-associated lipoprotein Alp-mRNA were significantly upregulated and the mRNA levels of factor H binding protein FhbA and immunogenic protein BipA were significantly downregulated in the spirochetes after acquisition into ticks in comparison with spirochetes grown in culture medium. In addition, genes such as bta124 and bta116 were significantly upregulated in spirochetes in unfed ticks in comparison with the levels noted in spirochetes after acquisition. These findings represent an efficient in vitro blood-feeding method to study B. turicatae gene expression after acquisition and colonization in these ticks. In summary, we report that B. turicatae survive and develop in the tick host when acquired by in vitro feeding. We also report that B. turicatae genes are differentially expressed in ticks in comparison with the in vitro-grown cultures, indicating influence of tick environment on spirochete gene expression.
Collapse
Affiliation(s)
- Girish Neelakanta
- Center for Molecular Medicine, College of Sciences, Old Dominion University, Norfolk, VA 23529 (; )
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529
| | - Hameeda Sultana
- Center for Molecular Medicine, College of Sciences, Old Dominion University, Norfolk, VA 23529 (; )
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529
| | - Daniel E Sonenshine
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529
| | - Richard T Marconi
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298
| |
Collapse
|
50
|
Cabezas-Cruz A, Alberdi P, Valdés JJ, Villar M, de la Fuente J. Anaplasma phagocytophilum Infection Subverts Carbohydrate Metabolic Pathways in the Tick Vector, Ixodes scapularis. Front Cell Infect Microbiol 2017; 7:23. [PMID: 28229048 PMCID: PMC5293764 DOI: 10.3389/fcimb.2017.00023] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/18/2017] [Indexed: 12/24/2022] Open
Abstract
The obligate intracellular pathogen, Anaplasma phagocytophilum, is the causative agent of human, equine, and canine granulocytic anaplasmosis and tick-borne fever (TBF) in ruminants. A. phagocytophilum has become an emerging tick-borne pathogen in the United States, Europe, Africa, and Asia, with increasing numbers of infected people and animals every year. It has been recognized that intracellular pathogens manipulate host cell metabolic pathways to increase infection and transmission in both vertebrate and invertebrate hosts. However, our current knowledge on how A. phagocytophilum affect these processes in the tick vector, Ixodes scapularis is limited. In this study, a genome-wide search for components of major carbohydrate metabolic pathways was performed in I. scapularis ticks for which the genome was recently published. The enzymes involved in the seven major carbohydrate metabolic pathways glycolysis, gluconeogenesis, pentose phosphate, tricarboxylic acid cycle (TCA), glyceroneogenesis, and mitochondrial oxidative phosphorylation and β-oxidation were identified. Then, the available transcriptomics and proteomics data was used to characterize the mRNA and protein levels of I. scapularis major carbohydrate metabolic pathway components in response to A. phagocytophilum infection of tick tissues and cultured cells. The results showed that major carbohydrate metabolic pathways are conserved in ticks. A. phagocytophilum infection inhibits gluconeogenesis and mitochondrial metabolism, but increases the expression of glycolytic genes. A model was proposed to explain how A. phagocytophilum could simultaneously control tick cell glucose metabolism and cytoskeleton organization, which may be achieved in part by up-regulating and stabilizing hypoxia inducible factor 1 alpha in a hypoxia-independent manner. The present work provides a more comprehensive view of the major carbohydrate metabolic pathways involved in the response to A. phagocytophilum infection in ticks, and provides the basis for further studies to develop novel strategies for the control of granulocytic anaplasmosis.
Collapse
Affiliation(s)
- Alejandro Cabezas-Cruz
- Institute of Parasitology, Biology Center, Czech Academy of SciencesCeské Budejovice, Czechia
- Faculty of Science, University of South BohemiaCeské Budejovice, Czechia
| | - Pilar Alberdi
- SaBio. Instituto de Investigación en Recursos Cinegéticos (CSIC-UCLM-JCCM)Ciudad Real, Spain
| | - James J. Valdés
- Institute of Parasitology, Biology Center, Czech Academy of SciencesCeské Budejovice, Czechia
- Department of Virology, Veterinary Research InstituteBrno, Czechia
| | - Margarita Villar
- SaBio. Instituto de Investigación en Recursos Cinegéticos (CSIC-UCLM-JCCM)Ciudad Real, Spain
| | - José de la Fuente
- SaBio. Instituto de Investigación en Recursos Cinegéticos (CSIC-UCLM-JCCM)Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State UniversityStillwater, OK, USA
| |
Collapse
|