1
|
Minayo Martín S, Villar M, Sánchez-Cano A, Fontoura-Gonçalves C, Hernández JM, Williams RAJ, Quevedo MÁ, Höfle U. Impact of urbanization on the house sparrow (Passer domesticus): Serum proteome and pathogen prevalence. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 968:178920. [PMID: 39987830 DOI: 10.1016/j.scitotenv.2025.178920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
The house sparrow (Passer domesticus) is a globally distributed species found in rural, urban and other humanised environments. In Europe, sparrow populations have significantly declined in recent decades, especially in urbanised areas. In the present study, we analysed the impact of urbanization on sparrow body condition, pathogen prevalence, and serum proteome changes. Sparrows were captured in four locations with two different urbanization status (rural/urban). Biometric data, blood samples and oral and cloacal swabs were collected. Rural sparrows exhibited significantly better body condition compared to urban sparrows, with no notable differences between sexes. Haemoparasite prevalence was higher in rural sparrows 70.16 % (87/124) than in urban sparrows 50 % (27/54). No avian influenza virus (AIV) or West Nile virus (WNV) genetic material was found, although one urban sparrow (0.58 %) had antibodies to AIV. Serum proteomics revealed that rural sparrows showed an up-regulation of proteins involved in the metabolism, in contrast to proteins of the immune system and the coagulation system, which were found to be over-represented in urban sparrows. Thus, we documented a worse body condition and immune system activation in urban sparrows in contrast to a more active metabolism and a higher prevalence of avian malaria in rural sparrows, and at least occasional exposure to AIV in urban habitats. This information suggests exposure to urban environments may alter the host-pathogen relationship. Urbanization in combination with exposure to AIV, could modulate their role in viral spread and transmission.
Collapse
Affiliation(s)
- Sara Minayo Martín
- SaBio Research Group, Institute for Game and Wildlife Research IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain.
| | - Margarita Villar
- SaBio Research Group, Institute for Game and Wildlife Research IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain; Biochemistry Section, Faculty of Sciences and Chemical Technologies, University of Castilla-La Mancha, Avda. Camilo Jose Cela 10, 13071 Ciudad Real, Spain
| | - Alberto Sánchez-Cano
- SaBio Research Group, Institute for Game and Wildlife Research IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain
| | - Catarina Fontoura-Gonçalves
- SaBio Research Group, Institute for Game and Wildlife Research IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain; CIBIO- Centro de Investigação em Biodiversidade e Recursos Genéticos. Rua Padre Armando Quintas, N° 7, Vairão 4485-661, Portugal; BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Campus de Vairão, 4485-661 Vairão, Portugal
| | - José Manuel Hernández
- SaBio Research Group, Institute for Game and Wildlife Research IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain
| | - Richard A J Williams
- Department of Genetics, Physiology and Microbiology, Faculty of Biology, Complutense University of Madrid (UCM), Calle Antonio Nováis 12, Ciudad Universitaria, 28040 Madrid, Spain
| | - Miguel Ángel Quevedo
- Centro de Conservación de la Biodiversidad Zoobotánico Jerez. Calle Madreselva, s/n, 11408, Jerez de la Frontera, Spain
| | - Ursula Höfle
- SaBio Research Group, Institute for Game and Wildlife Research IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain.
| |
Collapse
|
2
|
Zhang D, Yu L, Tang H, Niu H. Anaplasma phagocytophilum AFAP targets the host nucleolus and inhibits induced apoptosis. Front Microbiol 2025; 15:1533640. [PMID: 39839117 PMCID: PMC11747512 DOI: 10.3389/fmicb.2024.1533640] [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: 11/24/2024] [Accepted: 12/19/2024] [Indexed: 01/23/2025] Open
Abstract
Anaplasma phagocytophilum, the etiologic agent of human granulocytic anaplasmosis (HGA), is an obligate intracellular Gram-negative bacterium. During infection, A. phagocytophilum transfers its type IV secretion system (T4SS) effector proteins into host cells to manipulate cellular processes. AFAP (an actin filament-associated Anaplasma phagocytophilum protein) was identified as a T4SS effector protein and found to interact with the host nucleolin, as described in a previous study. In this study, proteomic analysis was performed to extensively identify AFAP-interacting proteins in host cells and analyze the potential role of AFAP in modulating host cellular processes. A total of 586 host proteins were identified interacting with AFAP by data-independent acquisition mass spectrometry and annotated to 501 Gene Ontology (GO) terms, with the significantly over-represented ones related to ribosomes, nucleolus, DNA binding, and rRNA metabolic process. Given the role of the nucleolus in cellular stress response, the targeting of AFAP to the nucleolus, and the identification of dozens of AFAP-interacting proteins that were annotated to the GO term (GO:0072331, signal transduction by p53 class mediator), the role of AFAP in modulating host apoptosis was determined. AFAP was found capable of inhibiting induced apoptosis. Thus, the proteomic analysis of AFAP-interacting proteins and determination of AFAP with anti-apoptotic activity may help elucidate the role of this T4SS effector protein in HGA pathogenesis.
Collapse
Affiliation(s)
- Daxiu Zhang
- Clinical Laboratory Center, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Lifeng Yu
- Clinical Laboratory Center, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Hui Tang
- Clinical Laboratory Center, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Hua Niu
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
3
|
Contreras M, Sobrino I, de la Fuente J. Paratransgenic quantum vaccinology. Trends Parasitol 2024; 40:1107-1114. [PMID: 39462754 DOI: 10.1016/j.pt.2024.10.006] [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: 09/26/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/29/2024]
Abstract
Tick vaccines are an environmentally friendly intervention for the prevention and control of tick-borne diseases affecting humans and animals worldwide. From our perspective, the challenges in tick vaccinology have encouraged the implementation of new interventions. In this opinion article we propose paratransgenic quantum vaccinology as a new approach that integrates platform trends in biotechnology, such as omics datasets combined with big data analytics, machine learning, and paratransgenesis with a systems biology perspective. This innovative approach allows the identification of protective epitopes in tick- and/or pathogen-derived proteins for the design of chimeric vaccine candidate antigens which can be produced by commensal/symbiotic microorganisms eliciting a protective response in the host.
Collapse
Affiliation(s)
- Marinela Contreras
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain.
| | - Isidro Sobrino
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - José de la Fuente
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain; Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA.
| |
Collapse
|
4
|
Hekimoğlu O, Sağlam İK. High Crimean-Congo hemorrhagic fever incidence linked to greater genetic diversity and differentiation in Hyalomma marginatum populations in Türkiye. Parasit Vectors 2024; 17:477. [PMID: 39587660 PMCID: PMC11590318 DOI: 10.1186/s13071-024-06530-z] [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: 08/13/2024] [Accepted: 10/08/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Ticks are crucial vectors of a wide range of pathogens, posing significant threats to human and animal health globally. Understanding the genetic basis of tick biology and host-parasite interactions is essential for developing effective control programs. This study investigates the fine-scale genetic structure of Hyalomma marginatum Koch, 1844, the primary vector of Crimean-Congo hemorrhagic fever (CCHF) in Türkiye. Despite its significant public health importance, information regarding its population structure and genetic diversity is quite limited. METHODS We used restriction site-associated DNA sequencing (RAD-Seq) to obtain genome-wide sequence data from 10 tick populations in Türkiye, collected from regions with low, moderate, and high incidence rates of CCHF. Based on these data, we determined population structure and diversity of populations using principal component analysis (PCA) and admixture analysis. Furthermore, we calculated pairwise FST and utilized discriminant analysis of principal components (DAPC) to understand genetic differentiation between populations. RESULTS PCA and admixture analysis indicated minimal genetic structure between populations, but we detected notable genetic differentiation and high genetic diversity from regions with high CCHF rates. Furthermore, our DAPC identified 31 significant single-nucleotide polymorphisms (SNPs) associated with regions with high CCHF incidence, with 25 SNPs located near genes involved in critical biological functions such as nucleic acid binding, transmembrane transport, and proteolysis. These findings suggest that genetic variations in these regions may confer adaptive advantages in environments with high pathogen loads. CONCLUSIONS This study provides the first comprehensive analysis of H. marginatum genetic diversity in Türkiye, revealing significant differentiation in populations from CCHF-endemic regions. These results underscore the importance of considering fine-scale genetic diversity to fully understand the drivers of genetic variation in ticks and their implications for vectorial capacity.
Collapse
Affiliation(s)
- Olcay Hekimoğlu
- Faculty of Science, Department of Biology, Division of Ecology, Hacettepe University, 06800, Beytepe, Ankara, Türkiye.
| | - İsmail K Sağlam
- Faculty of Science, Department of Molecular Biology and Genetics, Koc University, 34450, Ýstanbul, Türkiye
| |
Collapse
|
5
|
Maitre A, Kratou M, Corona-Guerrero I, Abuin-Denis L, Mateos-Hernández L, Mosqueda J, Almazan C, Said MB, Piloto-Sardiñas E, Obregon D, Cabezas-Cruz A. Differential interactions of Rickettsia species with tick microbiota in Rh. sanguineus and Rh. turanicus. Sci Rep 2024; 14:20674. [PMID: 39237587 PMCID: PMC11377539 DOI: 10.1038/s41598-024-71539-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024] Open
Abstract
Tick-borne rickettsioses, caused by Gram-negative bacteria of the Rickettsia genus, pose a growing global threat, with various arthropod vectors contributing to their transmission. Understanding the complex interactions within tick microbiota, including the role of Rickettsia species, is crucial for elucidating the dynamics of rickettsial diseases. Here, we investigate the taxonomic profiles and co-occurrence networks of Rickettsia in Rh. sanguineus sensus lato (s.l.) and Rh. turanicus ticks, revealing significant differences in community composition and local connectivity of Rickettsia species. While the microbiota of both tick species share common taxa, distinct differences in relative abundance and network topology suggest unique ecological niches. Moreover, robustness analysis demonstrates varying resilience to perturbations, indicating different strategies for network organization. Our findings also highlight metabolic differences between tick species, suggesting potential implications for Rickettsia interactions. Overall, this study provides insights into the intricate microbial landscape within ticks, shedding light on the functional redundancy and metabolic pathways associated with Rickettsia, thus advancing our understanding of tick-borne diseases.
Collapse
Affiliation(s)
- Apolline Maitre
- ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700, Maisons-Alfort, France
- INRAE, UR 0045 Laboratoire de Recherches sur le Développement de l'Elevage (SELMET-LRDE), 20250, Corte, France
- EA 7310, Laboratoire de Virologie, Université de Corse, Corte, France
| | - Myriam Kratou
- Laboratory of Microbiology, National School of Veterinary Medicine of Sidi Thabet, University of Manouba, 2010, Manouba, Tunisia
| | - Ivan Corona-Guerrero
- Immunology and Vaccines Laboratory, C. A. Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Queretaro, Mexico
- C.A. Salud Animal y Microbiologia Ambiental. Facultad de Ciencias Naturales, Universidad Autonoma de Queretaro, Queretaro, Mexico
| | - Lianet Abuin-Denis
- ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700, Maisons-Alfort, France
- Animal Biotechnology Department, Center for Genetic Engineering and Biotechnology, Avenue 31 between 158 and 190, P.O. Box 6162, 10600, Havana, Cuba
| | - Lourdes Mateos-Hernández
- ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700, Maisons-Alfort, France
| | - Juan Mosqueda
- Immunology and Vaccines Laboratory, C. A. Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Queretaro, Mexico
- C.A. Salud Animal y Microbiologia Ambiental. Facultad de Ciencias Naturales, Universidad Autonoma de Queretaro, Queretaro, Mexico
| | - Consuelo Almazan
- Immunology and Vaccines Laboratory, C. A. Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Queretaro, Mexico
- C.A. Salud Animal y Microbiologia Ambiental. Facultad de Ciencias Naturales, Universidad Autonoma de Queretaro, Queretaro, Mexico
| | - Mourad Ben Said
- Laboratory of Microbiology, National School of Veterinary Medicine of Sidi Thabet, University of Manouba, 2010, Manouba, Tunisia
- Department of Basic Sciences, Higher Institute of Biotechnology of Sidi Thabet, University of Manouba, 2010, Manouba, Tunisia
| | - Elianne Piloto-Sardiñas
- ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700, Maisons-Alfort, France
- Direction of Animal Health, National Center for Animal and Plant Health, Carretera de Tapaste y Autopista Nacional, Apartado Postal 10, 32700, San José de Las Lajas, Mayabeque, Cuba
| | - Dasiel Obregon
- School of Environmental Sciences, University of Guelph, Guelph, ON, Canada.
| | - Alejandro Cabezas-Cruz
- ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700, Maisons-Alfort, France.
| |
Collapse
|
6
|
Samaddar S, Rolandelli A, O'Neal AJ, Laukaitis-Yousey HJ, Marnin L, Singh N, Wang X, Butler LR, Rangghran P, Kitsou C, Cabrera Paz FE, Valencia L, R Ferraz C, Munderloh UG, Khoo B, Cull B, Rosche KL, Shaw DK, Oliver J, Narasimhan S, Fikrig E, Pal U, Fiskum GM, Polster BM, Pedra JHF. Bacterial reprogramming of tick metabolism impacts vector fitness and susceptibility to infection. Nat Microbiol 2024; 9:2278-2291. [PMID: 38997520 DOI: 10.1038/s41564-024-01756-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/11/2024] [Indexed: 07/14/2024]
Abstract
Arthropod-borne pathogens are responsible for hundreds of millions of infections in humans each year. The blacklegged tick, Ixodes scapularis, is the predominant arthropod vector in the United States and is responsible for transmitting several human pathogens, including the Lyme disease spirochete Borrelia burgdorferi and the obligate intracellular rickettsial bacterium Anaplasma phagocytophilum, which causes human granulocytic anaplasmosis. However, tick metabolic response to microbes and whether metabolite allocation occurs upon infection remain unknown. Here we investigated metabolic reprogramming in the tick ectoparasite I. scapularis and determined that the rickettsial bacterium A. phagocytophilum and the spirochete B. burgdorferi induced glycolysis in tick cells. Surprisingly, the endosymbiont Rickettsia buchneri had a minimal effect on bioenergetics. An unbiased metabolomics approach following A. phagocytophilum infection of tick cells showed alterations in carbohydrate, lipid, nucleotide and protein metabolism, including elevated levels of the pleiotropic metabolite β-aminoisobutyric acid. We manipulated the expression of genes associated with β-aminoisobutyric acid metabolism in I. scapularis, resulting in feeding impairment, diminished survival and reduced bacterial acquisition post haematophagy. Collectively, we discovered that metabolic reprogramming affects interspecies relationships and fitness in the clinically relevant tick I. scapularis.
Collapse
Affiliation(s)
- Sourabh Samaddar
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Agustin Rolandelli
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Anya J O'Neal
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hanna J Laukaitis-Yousey
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Liron Marnin
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Nisha Singh
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Department of Biotechnology, School of Energy Technology, Pandit Deendayal Energy University; Knowledge Corridor, Gandhinagar, India
| | - Xiaowei Wang
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
- MP Biomedicals, Solon, OH, USA
| | - L Rainer Butler
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Parisa Rangghran
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Chrysoula Kitsou
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Francy E Cabrera Paz
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Luisa Valencia
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Camila R Ferraz
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | | | - Benedict Khoo
- Division of Environmental Health Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Benjamin Cull
- Department of Entomology, University of Minnesota, Saint Paul, MN, USA
| | - Kristin L Rosche
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Dana K Shaw
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Jonathan Oliver
- Division of Environmental Health Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Sukanya Narasimhan
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Erol Fikrig
- Department of Internal Medicine, Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Gary M Fiskum
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Brian M Polster
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Bo Y, Yu Q, Gao W. Progress of depression mechanism based on Omics method. J Pharm Biomed Anal 2024; 240:115884. [PMID: 38183729 DOI: 10.1016/j.jpba.2023.115884] [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: 06/10/2023] [Revised: 11/24/2023] [Accepted: 11/26/2023] [Indexed: 01/08/2024]
Abstract
Depression is a very common disabling mental disorder, which is typically characterized by high rates of disability and mortality. Although research into the various mechanisms of depression was still underway, its physiopathology remains uncertain. The rapid developments in new technologies and the combined use of a variety of techniques will help to understand the pathogenesis of depression and explore effective treatment methods. In this review, we focus on the combination of proteomic and metabolomic approaches to analyze metabolites and proteins in animal models of depression induced by different modeling approaches, with the aim of broadening the understanding of the physiopathological mechanisms of depression using complementary "omics" strategy.
Collapse
Affiliation(s)
- Yaping Bo
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Qing Yu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Wenyuan Gao
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China.
| |
Collapse
|
10
|
Le Dortz LL, Rouxel C, Leroy Q, Ducongé F, Boulouis HJ, Haddad N, Deshuillers PL, Lagrée AC. Aptamer selection against cell extracts containing the zoonotic obligate intracellular bacterium, Anaplasma phagocytophilum. Sci Rep 2024; 14:2465. [PMID: 38291133 PMCID: PMC10828505 DOI: 10.1038/s41598-024-52808-8] [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: 11/14/2023] [Accepted: 01/23/2024] [Indexed: 02/01/2024] Open
Abstract
A. phagocytophilum is a zoonotic and tick-borne bacterium, threatening human and animal health. Many questions persist concerning the variability of strains and the mechanisms governing the interactions with its different hosts. These gaps can be explained by the difficulty to cultivate and study A. phagocytophilum because of its strict intracellular location and the lack of specific tools, in particular monoclonal antibodies, currently unavailable. The objective of our study was to develop DNA aptamers against A. phagocytophilum, or molecules expressed during the infection, as new study and/or capture tools. Selecting aptamers was a major challenge due to the strict intracellular location of the bacterium. To meet this challenge, we set up a customized selection protocol against an enriched suspension of A. phagocytophilum NY18 strain, cultivated in HL-60 cells. The implementation of SELEX allowed the selection of three aptamers, characterized by a high affinity for HL-60 cells infected with A. phagocytophilum NY18 strain. Interestingly, the targets of these three aptamers are most likely proteins expressed at different times of infection. The selected aptamers could contribute to increase our understanding of the interactions between A. phagocytophilum and its hosts, as well as permit the development of new diagnostic, therapeutic or drug delivery appliances.
Collapse
Affiliation(s)
- Lisa Lucie Le Dortz
- Anses, INRAe, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratory of Animal Health, 94700, Maisons-Alfort, France.
| | - Clotilde Rouxel
- Anses, INRAe, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratory of Animal Health, 94700, Maisons-Alfort, France
| | - Quentin Leroy
- Anses, INRAe, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratory of Animal Health, 94700, Maisons-Alfort, France
| | - Frédéric Ducongé
- CEA, Fundamental Research Division (DRF), Institute of Biology François Jacob, Molecular Imaging Research Center, CNRS UMR9199, Paris-Saclay University, 92265, Fontenay-Aux-Roses, France
| | - Henri-Jean Boulouis
- Anses, INRAe, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratory of Animal Health, 94700, Maisons-Alfort, France
| | - Nadia Haddad
- Anses, INRAe, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratory of Animal Health, 94700, Maisons-Alfort, France.
| | - Pierre Lucien Deshuillers
- Anses, INRAe, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratory of Animal Health, 94700, Maisons-Alfort, France
| | - Anne-Claire Lagrée
- Anses, INRAe, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratory of Animal Health, 94700, Maisons-Alfort, France
| |
Collapse
|
11
|
Vaz-Rodrigues R, Mazuecos L, Villar M, Contreras M, Artigas-Jerónimo S, González-García A, Gortázar C, de la Fuente J. Multi-omics analysis of zebrafish response to tick saliva reveals biological processes associated with alpha-Gal syndrome. Biomed Pharmacother 2023; 168:115829. [PMID: 37922649 DOI: 10.1016/j.biopha.2023.115829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/17/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023] Open
Abstract
The alpha-Gal syndrome (AGS) is a tick-borne allergy. A multi-omics approach was used to determine the effect of tick saliva and mammalian meat consumption on zebrafish gut transcriptome and proteome. Bioinformatics analysis using R software was focused on significant biological and metabolic pathway changes associated with AGS. Ortholog mapping identified highly concordant human ortholog genes for the detection of disease-enriched pathways. Tick saliva treatment increased zebrafish mortality, incidence of hemorrhagic type allergic reactions and changes in behavior and feeding patterns. Transcriptomics analysis showed downregulation of biological and metabolic pathways correlated with anti-alpha-Gal IgE and allergic reactions to tick saliva affecting blood circulation, cardiac and vascular smooth muscle contraction, behavior and sensory perception. Disease enrichment analysis revealed downregulated orthologous genes associated with human disorders affecting nervous, musculoskeletal, and cardiovascular systems. Proteomics analysis revealed suppression of pathways associated with immune system production of reactive oxygen species and cardiac muscle contraction. Underrepresented proteins were mainly linked to nervous and metabolic human disorders. Multi-omics data revealed inhibition of pathways associated with adrenergic signaling in cardiomyocytes, and heart and muscle contraction. Results identify tick saliva-related biological pathways supporting multisystemic organ involvement and linking α-Gal sensitization with other illnesses for the identification of potential disease biomarkers.
Collapse
Affiliation(s)
- Rita Vaz-Rodrigues
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Lorena Mazuecos
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Margarita Villar
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain; Biochemistry Section, Faculty of Science and Chemical Technologies, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - Marinela Contreras
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Sara Artigas-Jerónimo
- Biochemistry Section, Faculty of Science and Chemical Technologies, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - Almudena González-García
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Christian Gortázar
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain; Department of Veterinary Pathobiology, Centre for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA.
| |
Collapse
|
12
|
Rosche KL, Hurtado J, Fisk EA, Vosbigian KA, Warren AL, Sidak-Loftis LC, Wright SJ, Ramirez-Zepp E, Park JM, Shaw DK. PERK-mediated antioxidant response is key for pathogen persistence in ticks. mSphere 2023; 8:e0032123. [PMID: 37733353 PMCID: PMC10597351 DOI: 10.1128/msphere.00321-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 07/31/2023] [Indexed: 09/22/2023] Open
Abstract
A crucial phase in the life cycle of tick-borne pathogens is the time spent colonizing and persisting within the arthropod. Tick immunity is emerging as a key force shaping how transmissible pathogens interact with the vector. How pathogens remain in the tick despite immunological pressure remains unknown. In persistently infected Ixodes scapularis, we found that Borrelia burgdorferi (causative agent of Lyme disease) and Anaplasma phagocytophilum (causative agent of granulocytic anaplasmosis) activate a cellular stress pathway mediated by the endoplasmic reticulum receptor PKR-like ER kinase (PERK) and the central regulatory molecule eIF2α. Disabling the PERK pathway through pharmacological inhibition and RNA interference (RNAi) significantly decreased microbial numbers. In vivo RNAi of the PERK pathway not only reduced the number of A. phagocytophilum and B. burgdorferi colonizing larvae after a bloodmeal but also significantly reduced the number of bacteria that survive the molt. An investigation into PERK pathway-regulated targets revealed that A. phagocytophilum and B. burgdorferi induce activity of the antioxidant response regulator, nuclear factor erythroid 2-related factor 2 (Nrf2). Tick cells deficient for nrf2 expression or PERK signaling showed accumulation of reactive oxygen and nitrogen species in addition to reduced microbial survival. Supplementation with antioxidants rescued the microbicidal phenotype caused by blocking the PERK pathway. Altogether, our study demonstrates that the Ixodes PERK pathway is activated by transmissible microbes and facilitates persistence in the arthropod by potentiating an Nrf2-regulated antioxidant environment. IMPORTANCE Recent advances demonstrate that the tick immune system recognizes and limits the pathogens they transmit. Innate immune mediators such as antimicrobial peptides and reactive oxygen/nitrogen species are produced and restrict microbial survival. It is currently unclear how pathogens remain in the tick, despite this immune assault. We found that an antioxidant response controlled by the PERK branch of the unfolded protein response is activated in ticks that are persistently infected with Borrelia burgdorferi (Lyme disease) or Anaplasma phagocytophilum (granulocytic anaplasmosis). The PERK pathway induces the antioxidant response transcription factor, Nrf2, which coordinates a gene network that ultimately neutralizes reactive oxygen and nitrogen species. Interfering with this signaling cascade in ticks causes a significant decline in pathogen numbers. Given that innate immune products can cause collateral damage to host tissues, we speculate that this is an arthropod-driven response aimed at minimizing damage to "self" that also inadvertently benefits the pathogen. Collectively, our findings shed light on the mechanistic push and pull between tick immunity and pathogen persistence within the arthropod vector.
Collapse
Affiliation(s)
- Kristin L. Rosche
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Joanna Hurtado
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Elis A. Fisk
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Kaylee A. Vosbigian
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Ashley L. Warren
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Lindsay C. Sidak-Loftis
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Sarah J. Wright
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Elisabeth Ramirez-Zepp
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Jason M. Park
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Dana K. Shaw
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| |
Collapse
|
13
|
Panda M, Kalita E, Singh S, Rao A, Prajapati VK. Application of functional proteomics in understanding RNA virus-mediated infection. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 138:301-325. [PMID: 38220429 DOI: 10.1016/bs.apcsb.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Together with the expansion of genome sequencing research, the number of protein sequences whose function is yet unknown is increasing dramatically. The primary goals of functional proteomics, a developing area of study in the realm of proteomic science, are the elucidation of the biological function of unidentified proteins and the molecular description of cellular systems at the molecular level. RNA viruses have emerged as the cause of several human infectious diseases with large morbidity and fatality rates. The introduction of high-throughput sequencing tools and genetic-based screening approaches over the last few decades has enabled researchers to find previously unknown and perplexing elements of RNA virus replication and pathogenesis on a scale never feasible before. Viruses, on the other hand, frequently disrupt cellular proteostasis, macromolecular complex architecture or stoichiometry, and post-translational changes to take over essential host activities. Because of these consequences, structural and global protein and proteoform monitoring is highly necessiated. Mass spectrometry (MS) has the potential to elucidate key details of virus-host interactions and speed up the identification of antiviral targets, giving precise data on the stoichiometry of cellular and viral protein complexes as well as mechanistic insights, has lately emerged as a key part of the RNA virus biology toolbox as a functional proteomics approach. Affinity-based techniques are primarily employed to identify interacting proteins in stable complexes in living organisms. A protein's biological role is strongly suggested by its relationship with other members of a certain protein complex that is involved in a particular process. With a particular emphasis on the most recent advancements in defining host responses and their translational implications to uncover novel tractable antiviral targets, this chapter provides insight on several functional proteomics techniques in RNA virus biology.
Collapse
Affiliation(s)
- Mamta Panda
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India; Department of Neurology. Experimental Research in Stroke and Inflammation (ERSI),University Medical Center Hamburg-Eppendorf Martinistraße Hamburg, Germany
| | - Elora Kalita
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Satyendra Singh
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Abhishek Rao
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
14
|
Rosche KL, Hurtado J, Fisk EA, Vosbigian KA, Warren AL, Sidak-Loftis LC, Wright SJ, Ramirez-Zepp E, Park JM, Shaw DK. PERK-mediated antioxidant response is key for pathogen persistence in ticks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542958. [PMID: 37398437 PMCID: PMC10312570 DOI: 10.1101/2023.05.30.542958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
A crucial phase in the lifecycle of tick-borne pathogens is the time spent colonizing and persisting within the arthropod. Tick immunity is emerging as a key force shaping how transmissible pathogens interact with the vector. How pathogens remain in the tick despite immunological pressure remains unknown. In persistently infected Ixodes scapularis , we found that Borrelia burgdorferi (Lyme disease) and Anaplasma phagocytophilum (granulocytic anaplasmosis) activate a cellular stress pathway mediated by the endoplasmic reticulum receptor PERK and the central regulatory molecule, eIF2α. Disabling the PERK pathway through pharmacological inhibition and RNAi significantly decreased microbial numbers. In vivo RNA interference of the PERK pathway not only reduced the number of A. phagocytophilum and B. burgdorferi colonizing larvae after a bloodmeal, but also significantly reduced the number of bacteria that survive the molt. An investigation into PERK pathway-regulated targets revealed that A. phagocytophilum and B. burgdorferi induce activity of the antioxidant response regulator, Nrf2. Tick cells deficient for nrf2 expression or PERK signaling showed accumulation of reactive oxygen and nitrogen species in addition to reduced microbial survival. Supplementation with antioxidants rescued the microbicidal phenotype caused by blocking the PERK pathway. Altogether, our study demonstrates that the Ixodes PERK pathway is activated by transmissible microbes and facilitates persistence in the arthropod by potentiating an Nrf2-regulated antioxidant environment.
Collapse
Affiliation(s)
- Kristin L. Rosche
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Joanna Hurtado
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Elis A. Fisk
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Kaylee A. Vosbigian
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Ashley L. Warren
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Lindsay C. Sidak-Loftis
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Sarah J. Wright
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Elisabeth Ramirez-Zepp
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Jason M. Park
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
| | - Dana K. Shaw
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, USA
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| |
Collapse
|
15
|
Samaddar S, O'Neal AJ, Marnin L, Rolandelli A, Singh N, Wang X, Butler LR, Rangghran P, Laukaitis HJ, Cabrera Paz FE, Fiskum GM, Polster BM, Pedra JHF. Metabolic disruption impacts tick fitness and microbial relationships. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.26.542501. [PMID: 37292783 PMCID: PMC10245996 DOI: 10.1101/2023.05.26.542501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Arthropod-borne microbes rely on the metabolic state of a host to cycle between evolutionarily distant species. For instance, arthropod tolerance to infection may be due to redistribution of metabolic resources, often leading to microbial transmission to mammals. Conversely, metabolic alterations aids in pathogen elimination in humans, who do not ordinarily harbor arthropod-borne microbes. To ascertain the effect of metabolism on interspecies relationships, we engineered a system to evaluate glycolysis and oxidative phosphorylation in the tick Ixodes scapularis. Using a metabolic flux assay, we determined that the rickettsial bacterium Anaplasma phagocytophilum and the Lyme disease spirochete Borrelia burgdorferi, which are transstadially transmitted in nature, induced glycolysis in ticks. On the other hand, the endosymbiont Rickettsia buchneri, which is transovarially maintained, had a minimal effect on I. scapularis bioenergetics. Importantly, the metabolite β-aminoisobutyric acid (BAIBA) was elevated during A. phagocytophilum infection of tick cells following an unbiased metabolomics approach. Thus, we manipulated the expression of genes associated with the catabolism and anabolism of BAIBA in I. scapularis and detected impaired feeding on mammals, reduced bacterial acquisition, and decreased tick survival. Collectively, we reveal the importance of metabolism for tick-microbe relationships and unveil a valuable metabolite for I. scapularis fitness.
Collapse
Affiliation(s)
- Sourabh Samaddar
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - Anya J O'Neal
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - Liron Marnin
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - Agustin Rolandelli
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - Nisha Singh
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - Xiaowei Wang
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - L Rainer Butler
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - Parisa Rangghran
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Hanna J Laukaitis
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - Francy E Cabrera Paz
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| | - Gary M Fiskum
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Brian M Polster
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA
| |
Collapse
|
16
|
Mazuecos L, Alberdi P, Hernández-Jarguín A, Contreras M, Villar M, Cabezas-Cruz A, Simo L, González-García A, Díaz-Sánchez S, Neelakanta G, Bonnet SI, Fikrig E, de la Fuente J. Frankenbacteriosis targeting interactions between pathogen and symbiont to control infection in the tick vector. iScience 2023; 26:106697. [PMID: 37168564 PMCID: PMC10165458 DOI: 10.1016/j.isci.2023.106697] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/23/2023] [Accepted: 04/13/2023] [Indexed: 05/13/2023] Open
Abstract
Tick microbiota can be targeted for the control of tick-borne diseases such as human granulocytic anaplasmosis (HGA) caused by model pathogen, Anaplasma phagocytophilum. Frankenbacteriosis is inspired by Frankenstein and defined here as paratransgenesis of tick symbiotic/commensal bacteria to mimic and compete with tick-borne pathogens. Interactions between A. phagocytophilum and symbiotic Sphingomonas identified by metaproteomics analysis in Ixodes scapularis midgut showed competition between both bacteria. Consequently, Sphingomonas was selected for frankenbacteriosis for the control of A. phagocytophilum infection and transmission. The results showed that Franken Sphingomonas producing A. phagocytophilum major surface protein 4 (MSP4) mimic pathogen and reduce infection in ticks by competition and interaction with cell receptor components of infection. Franken Sphingomonas-MSP4 transovarial and trans-stadial transmission suggests that tick larvae with genetically modified Franken Sphingomonas-MSP4 could be produced in the laboratory and released in the field to compete and replace the wildtype populations with associated reduction in pathogen infection/transmission and HGA disease risks.
Collapse
Affiliation(s)
- Lorena Mazuecos
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Pilar Alberdi
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Angélica Hernández-Jarguín
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Marinela Contreras
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Margarita Villar
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Alejandro Cabezas-Cruz
- ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700 Maisons-Alfort, France
| | - Ladislav Simo
- ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, 94700 Maisons-Alfort, France
| | - Almudena González-García
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Sandra Díaz-Sánchez
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
| | - Girish Neelakanta
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
- Department of Biological Sciences, Old Dominion University, Norfolk, VA 23529, USA
| | - Sarah I. Bonnet
- Functional Genetics of Infectious Diseases Unit, Institut Pasteur, CNRS UMR 2000, Université de Paris, 75015 Paris, France
- Animal Health Department, INRAE, 37380 Nouzilly, France
| | - Erol Fikrig
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, CT 208022, USA
| | - José de la Fuente
- SaBio. Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA
- Corresponding author
| |
Collapse
|
17
|
Feng C, Torimaru K, Lim MYT, Chak LL, Shiimori M, Tsuji K, Tanaka T, Iida J, Okamura K. A novel eukaryotic RdRP-dependent small RNA pathway represses antiviral immunity by controlling an ERK pathway component in the black-legged tick. PLoS One 2023; 18:e0281195. [PMID: 36996253 PMCID: PMC10062562 DOI: 10.1371/journal.pone.0281195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 01/17/2023] [Indexed: 04/01/2023] Open
Abstract
Small regulatory RNAs (sRNAs) are involved in antiviral defense and gene regulation. Although roles of RNA-dependent RNA Polymerases (RdRPs) in sRNA biology are extensively studied in nematodes, plants and fungi, understanding of RdRP homologs in other animals is still lacking. Here, we study sRNAs in the ISE6 cell line, which is derived from the black-legged tick, an important vector of human and animal pathogens. We find abundant classes of ~22nt sRNAs that require specific combinations of RdRPs and sRNA effector proteins (Argonautes or AGOs). RdRP1-dependent sRNAs possess 5'-monophosphates and are mainly derived from RNA polymerase III-transcribed genes and repetitive elements. Knockdown of some RdRP homologs misregulates genes including RNAi-related genes and the regulator of immune response Dsor1. Sensor assays demonstrate that Dsor1 is downregulated by RdRP1 through the 3'UTR that contains a target site of RdRP1-dependent repeat-derived sRNAs. Consistent with viral gene repression by the RNAi mechanism using virus-derived small interfering RNAs, viral transcripts are upregulated by AGO knockdown. On the other hand, RdRP1 knockdown unexpectedly results in downregulation of viral transcripts. This effect is dependent on Dsor1, suggesting that antiviral immunity is enhanced by RdRP1 knockdown through Dsor1 upregulation. We propose that tick sRNA pathways control multiple aspects of immune response via RNAi and regulation of signaling pathways.
Collapse
Affiliation(s)
- Canran Feng
- Nara Institute of Science and Technology, Nara, Japan
| | | | - Mandy Yu Theng Lim
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Li-Ling Chak
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, Singapore
| | | | - Kosuke Tsuji
- Nara Institute of Science and Technology, Nara, Japan
| | - Tetsuya Tanaka
- Joint Faculty of Veterinary Medicine, Laboratory of Infectious Diseases, Kagoshima University, Kagoshima, Japan
| | - Junko Iida
- Nara Institute of Science and Technology, Nara, Japan
| | - Katsutomo Okamura
- Nara Institute of Science and Technology, Nara, Japan
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
18
|
De S, Kingan SB, Kitsou C, Portik DM, Foor SD, Frederick JC, Rana VS, Paulat NS, Ray DA, Wang Y, Glenn TC, Pal U. A high-quality Ixodes scapularis genome advances tick science. Nat Genet 2023; 55:301-311. [PMID: 36658436 DOI: 10.1038/s41588-022-01275-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 11/30/2022] [Indexed: 01/21/2023]
Abstract
Ixodes spp. and related ticks transmit prevalent infections, although knowledge of their biology and development of anti-tick measures have been hindered by the lack of a high-quality genome. In the present study, we present the assembly of a 2.23-Gb Ixodes scapularis genome by sequencing two haplotypes within one individual, complemented by chromosome-level scaffolding and full-length RNA isoform sequencing, yielding a fully reannotated genome featuring thousands of new protein-coding genes and various RNA species. Analyses of the repetitive DNA identified transposable elements, whereas the examination of tick-associated bacterial sequences yielded an improved Rickettsia buchneri genome. We demonstrate how the Ixodes genome advances tick science by contributing to new annotations, gene models and epigenetic functions, expansion of gene families, development of in-depth proteome catalogs and deciphering of genetic variations in wild ticks. Overall, we report critical genetic resources and biological insights impacting our understanding of tick biology and future interventions against tick-transmitted infections.
Collapse
Affiliation(s)
- Sandip De
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | | | - Chrysoula Kitsou
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | | | - Shelby D Foor
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Julia C Frederick
- Department of Environmental Health Science, University of Georgia, Athens, GA, USA
| | - Vipin S Rana
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Nicole S Paulat
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| | - David A Ray
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| | - Yan Wang
- Mass Spectrometry Facility, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Travis C Glenn
- Department of Environmental Health Science, University of Georgia, Athens, GA, USA.,Institute of Bioinformatics, University of Georgia, Athens, GA, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA. .,Virginia-Maryland College of Veterinary Medicine, College Park, MD, USA.
| |
Collapse
|
19
|
Filatov S, Dyčka F, Sterba J, Rego RO. A simple non-invasive method to collect soft tick saliva reveals differences in Ornithodoros moubata saliva composition between ticks infected and uninfected with Borrelia duttonii spirochetes. Front Cell Infect Microbiol 2023; 13:1112952. [PMID: 36743301 PMCID: PMC9895398 DOI: 10.3389/fcimb.2023.1112952] [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: 11/30/2022] [Accepted: 01/03/2023] [Indexed: 01/21/2023] Open
Abstract
Introduction: We developed a new simple method to assess the composition of proteinaceous components in the saliva of Ornithodoros moubata, the main vehicle for pathogen transmission and a likely source of bioactive molecules acting at the tick-vertebrate host interface. To collect naturally expectorated saliva from the ticks we employed an artificial membrane feeding technique using a simple, chemically defined diet containing phagostimulants and submitted native saliva samples collected in this way for liquid chromatography-mass spectrometry (LC-MS) analysis. These experiments were conducted with groups of uninfected ticks as well as with O. moubata infected with B. duttonii. The ticks exhibited a fair feeding response to the tested diet with engorgement rates reaching as high as 60-100% of ticks per feeding chamber. The LC-MS analysis identified a total of 17 and 15 proteins in saliva samples from the uninfected and infected O. moubata nymphs, respectively. Importantly, the analysis was sensitive enough to detect up to 9 different proteins in the samples of saliva containing diet upon which as few as 6 nymphal ticks fed during the experiments. Some of the proteins recognized in the analysis are well known for their immunomodulatory activity in a vertebrate host, whereas others are primarily thought of as structural or "housekeeping" proteins and their finding in the naturally expectorated tick saliva confirms that they can be secreted and might serve some functions at the tick-host interface. Most notably, some of the proteins that have long been suspected for their importance in the vector-pathogen interactions of Borrelia spirochetes were detected only in the samples from infected ticks, suggesting that their expression was altered by the persistent colonization of the tick's salivary glands by spirochetes. The simple method described herein is an important addition to the toolbox available to study the vector-host-pathogen interactions in the rapidly feeding soft ticks.
Collapse
Affiliation(s)
- Serhii Filatov
- National Scientific Center "Institute of Experimental and Clinical Veterinary Medicine", Kharkiv, Ukraine,*Correspondence: Serhii Filatov, ; Ryan O.M. Rego,
| | - Filip Dyčka
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czechia,Faculty of Science, University of South Bohemia, Ceske Budejovice, Czechia
| | - Jan Sterba
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czechia
| | - Ryan O.M. Rego
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czechia,Faculty of Science, University of South Bohemia, Ceske Budejovice, Czechia,*Correspondence: Serhii Filatov, ; Ryan O.M. Rego,
| |
Collapse
|
20
|
A systems biology approach to better understand human tick-borne diseases. Trends Parasitol 2023; 39:53-69. [PMID: 36400674 DOI: 10.1016/j.pt.2022.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 11/17/2022]
Abstract
Tick-borne diseases (TBDs) are a growing global health concern. Despite extensive studies, ill-defined tick-associated pathologies remain with unknown aetiologies. Human immunological responses after tick bite, and inter-individual variations of immune-response phenotypes, are not well characterised. Current reductive experimental methodologies limit our understanding of more complex tick-associated illness, which results from the interactions between the host, tick, and microbes. An unbiased, systems-level integration of clinical metadata and biological host data - obtained via transcriptomics, proteomics, and metabolomics - offers to drive the data-informed generation of testable hypotheses in TBDs. Advanced computational tools have rendered meaningful analysis of such large data sets feasible. This review highlights the advantages of integrative system biology approaches as essential for understanding the complex pathobiology of TBDs.
Collapse
|
21
|
Rana VS, Kitsou C, Dumler JS, Pal U. Immune evasion strategies of major tick-transmitted bacterial pathogens. Trends Microbiol 2023; 31:62-75. [PMID: 36055896 PMCID: PMC9772108 DOI: 10.1016/j.tim.2022.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/28/2022] [Accepted: 08/01/2022] [Indexed: 12/30/2022]
Abstract
Tick-transmitted bacterial pathogens thrive in enzootic infection cycles, colonizing disparate vertebrate and arthropod tissues, often establishing persistent infections. Therefore, the evolution of robust immune evasion strategies is central to their successful persistence or transmission between hosts. To survive in nature, these pathogens must counteract a broad range of microbicidal host responses that can be localized, tissue-specific, or systemic, including a mix of these responses at the host-vector interface. Herein, we review microbial immune evasion strategies focusing on Lyme disease spirochetes and rickettsial or tularemia agents as models for extracellular and intracellular tick-borne pathogens, respectively. A better understanding of these adaptive strategies could enrich our knowledge of the infection biology of relevant tick-borne diseases, contributing to the development of future preventions.
Collapse
Affiliation(s)
- Vipin Singh Rana
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - Chrysoula Kitsou
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA
| | - J Stephen Dumler
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD, USA.
| |
Collapse
|
22
|
Hodosi R, Kazimirova M, Soltys K. What do we know about the microbiome of I. ricinus? Front Cell Infect Microbiol 2022; 12:990889. [PMID: 36467722 PMCID: PMC9709289 DOI: 10.3389/fcimb.2022.990889] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 10/17/2022] [Indexed: 10/07/2023] Open
Abstract
I. ricinus is an obligate hematophagous parasitic arthropod that is responsible for the transmission of a wide range of zoonotic pathogens including spirochetes of the genus Borrelia, Rickettsia spp., C. burnetii, Anaplasma phagocytophilum and Francisella tularensis, which are part the tick´s microbiome. Most of the studies focus on "pathogens" and only very few elucidate the role of "non-pathogenic" symbiotic microorganisms in I. ricinus. While most of the members of the microbiome are leading an intracellular lifestyle, they are able to complement tick´s nutrition and stress response having a great impact on tick´s survival and transmission of pathogens. The composition of the tick´s microbiome is not consistent and can be tied to the environment, tick species, developmental stage, or specific organ or tissue. Ovarian tissue harbors a stable microbiome consisting mainly but not exclusively of endosymbiotic bacteria, while the microbiome of the digestive system is rather unstable, and together with salivary glands, is mostly comprised of pathogens. The most prevalent endosymbionts found in ticks are Rickettsia spp., Ricketsiella spp., Coxiella-like and Francisella-like endosymbionts, Spiroplasma spp. and Candidatus Midichloria spp. Since microorganisms can modify ticks' behavior, such as mobility, feeding or saliva production, which results in increased survival rates, we aimed to elucidate the potential, tight relationship, and interaction between bacteria of the I. ricinus microbiome. Here we show that endosymbionts including Coxiella-like spp., can provide I. ricinus with different types of vitamin B (B2, B6, B7, B9) essential for eukaryotic organisms. Furthermore, we hypothesize that survival of Wolbachia spp., or the bacterial pathogen A. phagocytophilum can be supported by the tick itself since coinfection with symbiotic Spiroplasma ixodetis provides I. ricinus with complete metabolic pathway of folate biosynthesis necessary for DNA synthesis and cell division. Manipulation of tick´s endosymbiotic microbiome could present a perspective way of I. ricinus control and regulation of spread of emerging bacterial pathogens.
Collapse
Affiliation(s)
- Richard Hodosi
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
| | - Maria Kazimirova
- Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Katarina Soltys
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
- Comenius University Science Park, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
23
|
Ambivalent Roles of Oxidative Stress in Triangular Relationships among Arthropod Vectors, Pathogens and Hosts. Antioxidants (Basel) 2022; 11:antiox11071254. [PMID: 35883744 PMCID: PMC9312350 DOI: 10.3390/antiox11071254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Blood-feeding arthropods, particularly ticks and mosquitoes are considered the most important vectors of arthropod-borne diseases affecting humans and animals. While feeding on blood meals, arthropods are exposed to high levels of reactive oxygen species (ROS) since heme and other blood components can induce oxidative stress. Different ROS have important roles in interactions among the pathogens, vectors, and hosts. ROS influence various metabolic processes of the arthropods and some have detrimental effects. In this review, we investigate the various roles of ROS in these arthropods, including their innate immunity and the homeostasis of their microbiomes, that is, how ROS are utilized to maintain the balance between the natural microbiota and potential pathogens. We elucidate the mechanism of how ROS are utilized to fight off invading pathogens and how the arthropod-borne pathogens use the arthropods’ antioxidant mechanism to defend against these ROS attacks and their possible impact on their vector potentials or their ability to acquire and transmit pathogens. In addition, we describe the possible roles of ROS in chemical insecticide/acaricide activity and/or in the development of resistance. Overall, this underscores the importance of the antioxidant system as a potential target for the control of arthropod and arthropod-borne pathogens.
Collapse
|
24
|
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
|
25
|
Gęgotek A, Moniuszko-Malinowska A, Groth M, Pancewicz S, Czupryna P, Dunaj J, Atalay S, Radziwon P, Skrzydlewska E. Plasma Proteomic Profile of Patients with Tick-Borne Encephalitis and Co-Infections. Int J Mol Sci 2022; 23:ijms23084374. [PMID: 35457192 PMCID: PMC9031133 DOI: 10.3390/ijms23084374] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/06/2022] [Accepted: 04/13/2022] [Indexed: 11/16/2022] Open
Abstract
Despite the increasing number of patients suffering from tick-borne encephalitis (TBE), Lyme disease, and their co-infection, the mechanisms of the development of these diseases and their effects on the human body are still unknown. Therefore, the aim of this study was to evaluate the changes in the proteomic profile of human plasma induced by the development of TBE and to compare it with changes in TBE patients co-infected with other tick-borne pathogens. The results obtained by proteomic analysis using a nanoLC-Q Exactive HF mass spectrometer showed that the most highly elevated groups of proteins in the plasma of TBE patients with co-infection were involved in the pro-inflammatory response and protein degradation, while the antioxidant proteins and factors responsible for protein biosynthesis were mainly downregulated. These results were accompanied by enhanced GSH- and 4-HNE-protein adducts formation, observed in TBE and co-infected patients at a higher level than in the case of patients with only TBE. In conclusion, the differences in the proteomic profiles between patients with TBE and co-infected patients indicate that these diseases are significantly diverse and, consequently, require different treatment, which is particularly important for further research, including the development of novel diagnostics tools.
Collapse
Affiliation(s)
- Agnieszka Gęgotek
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222 Bialystok, Poland; (S.A.); (E.S.)
- Correspondence: ; Tel.: +48-857485883
| | - Anna Moniuszko-Malinowska
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Zurawia 14, 15-540 Bialystok, Poland; (A.M.-M.); (M.G.); (S.P.); (P.C.); (J.D.)
| | - Monika Groth
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Zurawia 14, 15-540 Bialystok, Poland; (A.M.-M.); (M.G.); (S.P.); (P.C.); (J.D.)
| | - Sławomir Pancewicz
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Zurawia 14, 15-540 Bialystok, Poland; (A.M.-M.); (M.G.); (S.P.); (P.C.); (J.D.)
| | - Piotr Czupryna
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Zurawia 14, 15-540 Bialystok, Poland; (A.M.-M.); (M.G.); (S.P.); (P.C.); (J.D.)
| | - Justyna Dunaj
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Zurawia 14, 15-540 Bialystok, Poland; (A.M.-M.); (M.G.); (S.P.); (P.C.); (J.D.)
| | - Sinemyiz Atalay
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222 Bialystok, Poland; (S.A.); (E.S.)
| | - Piotr Radziwon
- Regional Centre for Transfusion Medicine, M. Sklodowskiej-Curie 23, 15-950 Bialystok, Poland;
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222 Bialystok, Poland; (S.A.); (E.S.)
| |
Collapse
|
26
|
Wang XR, Cull B. Apoptosis and Autophagy: Current Understanding in Tick–Pathogen Interactions. Front Cell Infect Microbiol 2022; 12:784430. [PMID: 35155277 PMCID: PMC8829008 DOI: 10.3389/fcimb.2022.784430] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/11/2022] [Indexed: 11/13/2022] Open
Abstract
Tick-borne diseases are a significant threat to human and animal health throughout the world. How tick-borne pathogens successfully infect and disseminate in both their vertebrate and invertebrate hosts is only partially understood. Pathogens have evolved several mechanisms to combat host defense systems, and to avoid and modulate host immunity during infection, therefore benefitting their survival and replication. In the host, pathogens trigger responses from innate and adaptive immune systems that recognize and eliminate invaders. Two important innate defenses against pathogens are the programmed cell death pathways of apoptosis and autophagy. This Mini Review surveys the current knowledge of apoptosis and autophagy pathways in tick-pathogen interactions, as well as the strategies evolved by pathogens for their benefit. We then assess the limitations to studying both pathways and discuss their participation in the network of the tick immune system, before highlighting future perspectives in this field. The knowledge gained would significantly enhance our understanding of the defense responses in vector ticks that regulate pathogen infection and burden, and form the foundation for future research to identify novel approaches to the control of tick-borne diseases.
Collapse
Affiliation(s)
- Xin-Ru Wang
- *Correspondence: Xin-Ru Wang, ; Benjamin Cull,
| | | |
Collapse
|
27
|
Villar M, Pacheco I, Mateos-Hernández L, Cabezas-Cruz A, Tabor AE, Rodríguez-Valle M, Mulenga A, Kocan KM, Blouin EF, de la Fuente J. Characterization of tick salivary gland and saliva alphagalactome reveals candidate alpha-gal syndrome disease biomarkers. Expert Rev Proteomics 2021; 18:1099-1116. [PMID: 34904495 DOI: 10.1080/14789450.2021.2018305] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Ticks are obligate hematophagous arthropods that synthesize the glycan Galα1-3Galβ1-(3)4GlcNAc-R (α-Gal) associated with the alpha-gal syndrome (AGS) or allergy to mammalian meat consumption. RESEARCH DESIGN AND METHODS In this study, we used a proteomics approach to characterize tick proteins in salivary glands (sialome SG), secreted saliva (sialome SA) and with α-Gal modification (alphagalactome SG and SA) in model tick species associated with the AGS in the United States (Amblyomma americanum) and Australia (Ixodes holocyclus). Selected proteins reactive to sera (IgE) from patients with AGS were identified to advance in the identification of possible proteins associated with the AGS. For comparative analysis, the α-Gal content was measured in various tick species. RESULTS The results confirmed that ticks produce proteins with α-Gal modifications and secreted into saliva during feeding. Proteins identified in tick alphagalactome SA by sera from patients with severe AGS symptomatology may constitute candidate disease biomarkers. CONCLUSIONS The results support the presence tick-derived proteins with α-Gal modifications in the saliva with potential implications in AGS and other disorders and protective capacity against tick infestations and pathogen infection. Future research should focus on the characterization of the function of tick glycoproteins with α-Gal in tick biology and AGS.
Collapse
Affiliation(s)
- Margarita Villar
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain.,Biochemistry Section, Faculty of Science and Chemical Technologies, and Regional Centre for Biomedical Research (CRIB), University of Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Iván Pacheco
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain
| | - Lourdes Mateos-Hernández
- UMR BIPAR, INRAE, ANSES, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, 94700, France
| | - Alejandro Cabezas-Cruz
- UMR BIPAR, INRAE, ANSES, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, 94700, France
| | - Ala E Tabor
- Queensland Alliance for Agriculture & Food Innovation, Centre for Animal Science, The University of Queensland, 306 Carmody Road, St. Lucia, QLD 4072, Australia.,School of Chemistry and Molecular Biosciences, The University of Queensland, 68 Cooper Road, St. Lucia, QLD 4072, Australia
| | - Manuel Rodríguez-Valle
- Queensland Alliance for Agriculture & Food Innovation, Centre for Animal Science, The University of Queensland, 306 Carmody Road, St. Lucia, QLD 4072, Australia
| | - Albert Mulenga
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX77843, United States
| | - Katherine M Kocan
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Edmour F Blouin
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain.,Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
28
|
Aguilar-Díaz H, Quiroz-Castañeda RE, Salazar-Morales K, Cossío-Bayúgar R, Miranda-Miranda E. Tick Immunobiology and Extracellular Traps: An Integrative Vision to Control of Vectors. Pathogens 2021; 10:pathogens10111511. [PMID: 34832666 PMCID: PMC8621429 DOI: 10.3390/pathogens10111511] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/01/2021] [Accepted: 11/16/2021] [Indexed: 01/21/2023] Open
Abstract
Ticks are hematophagous ectoparasites that infest a diverse number of vertebrate hosts. The tick immunobiology plays a significant role in establishing and transmitting many pathogens to their hosts. To control tick infestations, the acaricide application is a commonly used method with severe environmental consequences and the selection of tick-resistant populations. With these drawbacks, new tick control methods need to be developed, and the immune system of ticks contains a plethora of potential candidates for vaccine design. Additionally, tick immunity is based on an orchestrated action of humoral and cellular immune responses. Therefore, the actors of these responses are the object of our study in this review since they are new targets in anti-tick vaccine design. We present their role in the immune response that positions them as feasible targets that can be blocked, inhibited, interfered with, and overexpressed, and then elucidate a new method to control tick infestations through the development of vaccines. We also propose Extracellular Traps Formation (ETosis) in ticks as a process to eliminate their natural enemies and those pathogens they transmit (vectorial capacity), which results attractive since they are a source of acting molecules with potential use as vaccines.
Collapse
Affiliation(s)
- Hugo Aguilar-Díaz
- Unidad de Artropodología, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad INIFAP, Jiutepec 62574, Mexico; (R.C.-B.); (E.M.-M.)
- Correspondence:
| | - Rosa Estela Quiroz-Castañeda
- Unidad de Anaplasmosis, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad INIFAP, Jiutepec 62574, Mexico;
| | - Karina Salazar-Morales
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Mexico;
| | - Raquel Cossío-Bayúgar
- Unidad de Artropodología, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad INIFAP, Jiutepec 62574, Mexico; (R.C.-B.); (E.M.-M.)
| | - Estefan Miranda-Miranda
- Unidad de Artropodología, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad INIFAP, Jiutepec 62574, Mexico; (R.C.-B.); (E.M.-M.)
| |
Collapse
|
29
|
Huertas-Alonso AJ, Gavahian M, González-Serrano DJ, Hadidi M, Salgado-Ramos M, Sánchez-Verdú MP, Simirgiotis MJ, Barba FJ, Franco D, Lorenzo JM, Moreno A. Valorization of Wastewater from Table Olives: NMR Identification of Antioxidant Phenolic Fraction and Microwave Single-Phase Reaction of Sugary Fraction. Antioxidants (Basel) 2021; 10:antiox10111652. [PMID: 34829523 PMCID: PMC8615242 DOI: 10.3390/antiox10111652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 10/13/2021] [Accepted: 10/18/2021] [Indexed: 11/16/2022] Open
Abstract
The table olive industry is producing a huge amount of wastewater, which is a post-processing cost and an environmental concern. The present study aims to valorize this processing by-product to obtain a value-added product, thereby enhancing resource efficiency and contributing to achieving sustainable development goals (SDGs). In this sense, a chemical reaction-based platform was developed to obtain valuable components, such as levulinic acid (LA) and 5-hydromethylfurfural (HMF). The products were then analyzed using NMR identification of the antioxidant phenolic fraction and microwave single-phase reaction of the sugary fraction. According to the results, the highest concentration of phenolic compounds does not correspond to the sample directly obtained from NaOH treatment (S1), indicating that water washing steps (S2–S5) are fundamental to recover phenolic substances. Moreover, glucose was presented in the sugary fraction that can be transformed into levulinic acid by a single-phase reaction under microwave irradiation. The information provided in this manuscript suggests that the wastewater from the olive processing industry can be valorized to obtain valuable products.
Collapse
Affiliation(s)
- Alberto J. Huertas-Alonso
- Department of Organic Chemistry, Faculty of Chemical Sciences and Technologies (San Alberto Magno Building), University of Castilla-La Mancha, Av. Camilo José Cela, 10, 13071 Ciudad Real, Spain; (A.J.H.-A.); (D.J.G.-S.); (M.H.); (M.S.-R.); (M.P.S.-V.)
| | - Mohsen Gavahian
- Department of Food Science, National Pingtung University of Science and Technology, 1, Shuefu Road, Neipu, Pingtung 91201, Taiwan;
| | - Diego J. González-Serrano
- Department of Organic Chemistry, Faculty of Chemical Sciences and Technologies (San Alberto Magno Building), University of Castilla-La Mancha, Av. Camilo José Cela, 10, 13071 Ciudad Real, Spain; (A.J.H.-A.); (D.J.G.-S.); (M.H.); (M.S.-R.); (M.P.S.-V.)
| | - Milad Hadidi
- Department of Organic Chemistry, Faculty of Chemical Sciences and Technologies (San Alberto Magno Building), University of Castilla-La Mancha, Av. Camilo José Cela, 10, 13071 Ciudad Real, Spain; (A.J.H.-A.); (D.J.G.-S.); (M.H.); (M.S.-R.); (M.P.S.-V.)
| | - Manuel Salgado-Ramos
- Department of Organic Chemistry, Faculty of Chemical Sciences and Technologies (San Alberto Magno Building), University of Castilla-La Mancha, Av. Camilo José Cela, 10, 13071 Ciudad Real, Spain; (A.J.H.-A.); (D.J.G.-S.); (M.H.); (M.S.-R.); (M.P.S.-V.)
| | - M. Prado Sánchez-Verdú
- Department of Organic Chemistry, Faculty of Chemical Sciences and Technologies (San Alberto Magno Building), University of Castilla-La Mancha, Av. Camilo José Cela, 10, 13071 Ciudad Real, Spain; (A.J.H.-A.); (D.J.G.-S.); (M.H.); (M.S.-R.); (M.P.S.-V.)
| | - Mario J. Simirgiotis
- Institute of Pharmacy, Faculty of Sciences, Campus Isla Teja, Universidad Austral de Chile, Valdivia 5090000, Chile;
| | - Francisco J. Barba
- Nutrition and Food Science Area, Preventive Medicine and Public Health, Food Science, Toxicology and Forensic Medicine Department, Faculty of Pharmacy, Universitat de València, Av. Vicent Andrés Estellés, s/n, Burjassot, 46100 València, Spain;
| | - Daniel Franco
- Centro Tecnológico de la Carne de Galicia, Av. Galicia No. 4, Parque Tecnológico de Galicia, San Cibrao das Viñas, 32900 Ourense, Spain;
- Correspondence: (D.F.); (A.M.)
| | - José M. Lorenzo
- Centro Tecnológico de la Carne de Galicia, Av. Galicia No. 4, Parque Tecnológico de Galicia, San Cibrao das Viñas, 32900 Ourense, Spain;
- Área de Tecnología de los Alimentos, Facultad de Ciencias de Ourense, Universidad de Vigo, 32004 Ourense, Spain
| | - Andrés Moreno
- Department of Organic Chemistry, Faculty of Chemical Sciences and Technologies (San Alberto Magno Building), University of Castilla-La Mancha, Av. Camilo José Cela, 10, 13071 Ciudad Real, Spain; (A.J.H.-A.); (D.J.G.-S.); (M.H.); (M.S.-R.); (M.P.S.-V.)
- Correspondence: (D.F.); (A.M.)
| |
Collapse
|
30
|
Gomez-Chamorro A, Hodžić A, King KC, Cabezas-Cruz A. Ecological and evolutionary perspectives on tick-borne pathogen co-infections. CURRENT RESEARCH IN PARASITOLOGY & VECTOR-BORNE DISEASES 2021; 1:100049. [PMID: 35284886 PMCID: PMC8906131 DOI: 10.1016/j.crpvbd.2021.100049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/09/2021] [Accepted: 09/16/2021] [Indexed: 06/14/2023]
Abstract
Tick-borne pathogen co-infections are common in nature. Co-infecting pathogens interact with each other and the tick microbiome, which influences individual pathogen fitness, and ultimately shapes virulence, infectivity, and transmission. In this review, we discuss how tick-borne pathogens are an ideal framework to study the evolutionary dynamics of co-infections. We highlight the importance of inter-species and intra-species interactions in vector-borne pathogen ecology and evolution. We also propose experimental evolution in tick cell lines as a method to directly test the impact of co-infections on pathogen evolution. Experimental evolution can simulate in real-time the long periods of time involved in within-vector pathogen interactions in nature, a major practical obstacle to cracking the influence of co-infections on pathogen evolution and ecology.
Collapse
Affiliation(s)
- Andrea Gomez-Chamorro
- Department of Zoology, University of Oxford, Oxford, OX1 3SZ, UK
- Anses, INRAE, Ecole Nationale Vétérinaire D’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, F-94700, France
| | - Adnan Hodžić
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210, Vienna, Austria
| | - Kayla C. King
- Department of Zoology, University of Oxford, Oxford, OX1 3SZ, UK
| | - Alejandro Cabezas-Cruz
- Anses, INRAE, Ecole Nationale Vétérinaire D’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, F-94700, France
| |
Collapse
|
31
|
Aguilar-Díaz H, Quiroz-Castañeda RE, Cobaxin-Cárdenas M, Salinas-Estrella E, Amaro-Estrada I. Advances in the Study of the Tick Cattle Microbiota and the Influence on Vectorial Capacity. Front Vet Sci 2021; 8:710352. [PMID: 34485437 PMCID: PMC8415903 DOI: 10.3389/fvets.2021.710352] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 07/19/2021] [Indexed: 12/16/2022] Open
Abstract
The information from the tick cattle microbiota suggests that the microbial populations may modulate a successful infection process of the tick-borne pathogens. Therefore, there is a need to know the microbial population and their interactions. In this mini-review, we present several examples of how microbiota regulates the survival of pathogens inside the tick and contributes to fitness, adaptation, and tick immunity, among others. The communication between the tick microbiota and the host microbiota is vital to understanding the pathogen transmission process. As part of the tick microbiota, the pathogen interacts with different microbial populations, including the microorganisms of the host microbiota. These interactions comprise a microsystem that regulates the vectorial capacity involved in tick-borne diseases. The knowledge we have about the vectorial capacity contributes to a better understanding of tick-borne pathogens. Additionally, using approaches based on multi-omics strategies applied to studying the microbiota and its microbiome allows the development of strategies to control ticks. The results derived from those studies reveal the dynamics of the microbiota and potential targets for anti-tick vaccine development. In this context, the anti-microbiota vaccines have emerged as an alternative with a good prognosis. Some strategies developed to control other arthropods vectors, such as paratransgenesis, could control ticks and tick-borne diseases.
Collapse
Affiliation(s)
- Hugo Aguilar-Díaz
- Unidad de Artropodología del Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad, Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias, Morelos, Mexico
| | - Rosa Estela Quiroz-Castañeda
- Unidad de Anaplasmosis del Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad, Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias, Morelos, Mexico
| | - Mayra Cobaxin-Cárdenas
- Unidad de Anaplasmosis del Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad, Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias, Morelos, Mexico
| | - Elizabeth Salinas-Estrella
- Unidad de Anaplasmosis del Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad, Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias, Morelos, Mexico
| | - Itzel Amaro-Estrada
- Unidad de Anaplasmosis del Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad, Instituto Nacional de Investigaciones Forestales, Agrícolas y Pecuarias, Morelos, Mexico
| |
Collapse
|
32
|
Untargeted GC-TOFMS Analysis Reveals Metabolomic Changes in Salvia miltiorrhiza Bunge Leaf and Root in Response to Long-Term Drought Stress. HORTICULTURAE 2021. [DOI: 10.3390/horticulturae7070175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Salvia miltiorrhiza Bunge (Danshen) is an important traditional Chinese medicine herb. This study aimed to investigate the drought-responsive metabolic profiling in S. miltiorrhiza using gas chromatography time-of-flight mass spectrometry (GC-TOFMS) analysis. Fifty day-old S. miltiorrhiza seedlings were treated with two (moderate drought, MD) and four weeks (high drought, HD) of withholding water. The S. miltiorrhiza leaf and root samples were prepared for the GC-TOFMS analysis. Differential metabolites with substantial changes in content in S. miltiorrhiza leaf and root were identified using multivariate and univariate statistics. A total of 178 and 157 annotated metabolites were detected in S. miltiorrhiza leaf and root, respectively. Multivariate analysis showed that significantly discriminant metabolites in S. miltiorrhiza leaf by drought were associated with “galactose metabolism” and “citrate cycle”. In addition, the significantly discriminant metabolites in S. miltiorrhiza root were associated with “starch and sucrose metabolism”. Univariate statistics showed that the content of succinic acid, d-glucose, and oxoglutaric acid in S. miltiorrhiza leaf was increased by drought (fold change, FC > 1.5). Allose, d-xylose, melibiose, mannose, sorbitol, quinic acid, sinigrin, and taurine in S. miltiorrhiza root were decreased by drought (FC < 0.67). There were different metabolic profiles between S. miltiorrhiza leaf and root. However, the influence of drought stress on the pharmacological value and accumulation of bioactive constituents in S. miltiorrhiza should be further investigated.
Collapse
|
33
|
Ma Z, Li R, Hu R, Zheng W, Yu S, Cheng K, Zhang H, Xiao Y, Yi J, Wang Z, Wang Y, Chen C. Anaplasma phagocytophilum AptA enhances the UPS, autophagy, and anti-apoptosis of host cells by PSMG3. Int J Biol Macromol 2021; 184:497-508. [PMID: 34126152 DOI: 10.1016/j.ijbiomac.2021.06.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/03/2021] [Accepted: 06/06/2021] [Indexed: 12/12/2022]
Abstract
Anaplasma phagocytophilum is an obligate intracellular bacterium and a common tick-borne infectious pathogen that can cause human granulocytic anaplasmosis (HGA). Effector proteins play an important role in the pathogenic mechanism of A. phagocytophilum, but the specifics of the disease mechanism are unclear. We studied the effector protein AptA (A. phagocytophilum toxin A) using yeast two hybrid assays to screen its interacting protein proteasome assembly chaperone 3 (PSMG3, PAC3), and identified new mechanisms for the pathogenicity of A. phagocytophilum in HEK293T cells. After AptA enters the host cell, it interacts with PSMG3 to enhance the activity of the proteasome, causing ubiquitination and autophagy in the host cell and thereby increasing cross-talk between the ubiquitination-proteasome system (UPS) and autophagy. AptA also reduces the apoptotic efficiency of the host cells. These results offer new clues as to the pathogenic mechanism of A. phagocytophilum and support the hypothesis that AptA interacts with host PSMG3.
Collapse
Affiliation(s)
- Zhongchen Ma
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China; Collaborative Innovation Center for prevention and control of high incidence zoonotic infectious diseases in Western China, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China
| | - Ruirui Li
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China; Collaborative Innovation Center for prevention and control of high incidence zoonotic infectious diseases in Western China, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China
| | - Ruirui Hu
- College of Life Sciences, Shihezi University, 832003 Shihezi, Xinjiang, China
| | - Wei Zheng
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China; Collaborative Innovation Center for prevention and control of high incidence zoonotic infectious diseases in Western China, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China
| | - Shuifa Yu
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China; Collaborative Innovation Center for prevention and control of high incidence zoonotic infectious diseases in Western China, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China
| | - Kejian Cheng
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China; Collaborative Innovation Center for prevention and control of high incidence zoonotic infectious diseases in Western China, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China
| | - Huan Zhang
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China; Collaborative Innovation Center for prevention and control of high incidence zoonotic infectious diseases in Western China, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China
| | - Yangyang Xiao
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China; Collaborative Innovation Center for prevention and control of high incidence zoonotic infectious diseases in Western China, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China
| | - Jihai Yi
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China; Collaborative Innovation Center for prevention and control of high incidence zoonotic infectious diseases in Western China, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China
| | - Zhen Wang
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China; Collaborative Innovation Center for prevention and control of high incidence zoonotic infectious diseases in Western China, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China
| | - Yong Wang
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China; Collaborative Innovation Center for prevention and control of high incidence zoonotic infectious diseases in Western China, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China.
| | - Chuangfu Chen
- International Research Center for Animal Health Breeding, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China; Collaborative Innovation Center for prevention and control of high incidence zoonotic infectious diseases in Western China, College of Animal Science and Technology, Shihezi University, 832003 Shihezi, Xinjiang, China.
| |
Collapse
|
34
|
Talactac MR, Hernandez EP, Hatta T, Yoshii K, Kusakisako K, Tsuji N, Tanaka T. The antiviral immunity of ticks against transmitted viral pathogens. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 119:104012. [PMID: 33484780 DOI: 10.1016/j.dci.2021.104012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 06/12/2023]
Abstract
Ticks, being obligate hematophagous arthropods, are exposed to various blood-borne pathogens, including arboviruses. Consequently, their feeding behavior can readily transmit economically important viral pathogens to humans and animals. With this tightly knit vector and pathogen interaction, the replication and transmission of tick-borne viruses (TBVs) must be highly regulated by their respective tick vectors to avoid any adverse effect on the ticks' biological development and viability. Knowledge about the tick-virus interface, although gaining relevant advances in recent years, is advancing at a slower pace than the scientific developments related to mosquito-virus interactions. The unique and complicated feeding behavior of ticks, compared to that of other blood-feeding arthropods, also limits the studies that would further elaborate the antiviral immunity of ticks against TBVs. Hence, knowledge of molecular and cellular immune mechanisms at the tick-virus interface, will further elucidate the successful viral replication of TBVs in ticks and their effective transmission to human and animal hosts.
Collapse
Affiliation(s)
- Melbourne Rio Talactac
- Department of Clinical and Population Health, College of Veterinary Medicine and Biomedical Sciences, Cavite State University, Cavite, 4122, Philippines
| | - Emmanuel Pacia Hernandez
- Department of Parasitology and Tropical Medicine, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa, 252-0374, Japan
| | - Takeshi Hatta
- Department of Parasitology and Tropical Medicine, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa, 252-0374, Japan
| | - Kentaro Yoshii
- National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Kodai Kusakisako
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, 034-8628, Japan
| | - Naotoshi Tsuji
- Department of Parasitology and Tropical Medicine, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami, Sagamihara, Kanagawa, 252-0374, Japan
| | - Tetsuya Tanaka
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0065, Japan.
| |
Collapse
|
35
|
Screening of metabolites in the treatment of liver cancer xenografts HepG2/ADR by psoralen-loaded lipid nanoparticles. Eur J Pharm Biopharm 2021; 165:337-344. [PMID: 34062256 DOI: 10.1016/j.ejpb.2021.05.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Our study aimed to find potential biomarkers for drug resistance in liver cancer cells using metabolomics and further to evaluate the potential of psoralen-loaded polymer lipid nanoparticles (PSO-PLNs) to reverse the resistance of cells to doxorubicin. METHODS We used LC-MS-based non-targeted metabolomics, also known as global metabolite profiling, to screen in serum and urine of mice engrafted with a liver cancer cell line sensitive (HepG2/S) or resistant to doxorubicin (HepG2/ADR) for differentially regulated metabolites. We subsequently quantified the abundance of these metabolites in serum and the urine of mice. The mice were engrafted with HepG2 cells resistant against doxorubicin and were treated with I) doxorubicin, II) a combination of doxorubicin and psoralen and III) a combination of doxorubicin and psoralen packed in polymer lipid nanoparticles. RESULTS Metabolites found to be differentially present in urine of mice engrafted with resistant HepG2 cells were: hippuric acid, hyaluronic acid, pantothenic acid, and betaine; retinoic acid and α-linolenic acid were found to be reduced in serum samples of mice with HepG2 cells resistant to doxorubicin. The targeted analysis showed that the degree of regression of metabolic markers in groups differed: treatment group 2 had stronger degree of regression than treatment group 1 and the negative control group had the smallest, which indicates that the PSO-PLNs have superior properties compared with other treatments. CONCLUSION Psoralen reverses drug resistance of liver cancer cells and its efficacy can be increased by encapsulation in polymer lipid nanoparticles.
Collapse
|
36
|
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
|
37
|
Du Y, Mi Z, Xie Y, Lu D, Zheng H, Sun H, Zhang M, Niu Y. Insights into the molecular basis of tick-borne encephalitis from multiplatform metabolomics. PLoS Negl Trop Dis 2021; 15:e0009172. [PMID: 33690602 PMCID: PMC7984639 DOI: 10.1371/journal.pntd.0009172] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 03/22/2021] [Accepted: 01/23/2021] [Indexed: 12/30/2022] Open
Abstract
Background Tick-borne encephalitis virus (TBEV) is the most prevalent arbovirus, with a tentative estimate of 10,000 to 10,500 infections occurring in Europe and Asia every year. Endemic in Northeast China, tick-borne encephalitis (TBE) is emerging as a major threat to public health, local economies and tourism. The complicated array of host physiological changes has hampered elucidation of the molecular mechanisms underlying the pathogenesis of this disease. Methodology/Principle findings System-level characterization of the serum metabolome and lipidome of adult TBEV patients and a healthy control group was performed using liquid chromatography tandem mass spectrometry. By tracking metabolic and lipid changes during disease progression, crucial physiological changes that coincided with disease stages could be identified. Twenty-eight metabolites were significantly altered in the sera of TBE patients in our metabolomic analysis, and 14 lipids were significantly altered in our lipidomics study. Among these metabolites, alpha-linolenic acid, azelaic acid, D-glutamine, glucose-1-phosphate, L-glutamic acid, and mannose-6-phosphate were altered compared to the control group, and PC(38:7), PC(28:3;1), TAG(52:6), etc. were altered based on lipidomics. Major perturbed metabolic pathways included amino acid metabolism, lipid and oxidative stress metabolism (lipoprotein biosynthesis, arachidonic acid biosynthesis, leukotriene biosynthesis and sphingolipid metabolism), phospholipid metabolism and triglyceride metabolism. These metabolites were significantly perturbed during disease progression, implying their latent utility as prognostic markers. Conclusions/Significance TBEV infection causes distinct temporal changes in the serum metabolome and lipidome, and many metabolites are potentially involved in the acute inflammatory response and immune regulation. Our global analysis revealed anti- and pro-inflammatory processes in the host and changes to the entire metabolic profile. Relationships between metabolites and pathologies were established. This study provides important insight into the pathology of TBE, including its pathology, and lays the foundation for further research into putative markers of TBE disease. Tick-borne encephalitis virus (TBEV) with extreme contagiousness is a key danger to public health systems in Europe and Asia. To date, little information is obtained about the molecular mechanism underlying infection, and although commercial vaccines against TBEV exist, there is no specific treatment for the disease. Metabolomics and lipidomics offer multiple-visions of metabolome and lipidome sights and help elucidating metabolic to disease phenotype. Serum metabolism and lipidome analysis were performed based on mass spectrometer (MS) platform on a cohort of TBEV patients. About 400 metabolites performed crucial shifts in TBEV patients compared with healthy subjects. This study revealed that in the stage of infection, the host metabolome is tightly regulated, with anti-inflammatory processes modulating pro-inflammatory processes implying the self-limiting phenotype of TBEV and the inherent regulation in humans. The crucial perturbed metabolic pathways contained amino acid metabolism, fatty acid metabolism and phospholipid metabolism. This study provides a powerful and new approach to decipher the interactions between host and virus. These potential metabolites provide high sensitivity and specificity and have the capacity to function as biomarkers for disease surveillance and estimation of therapeutic interventions.
Collapse
Affiliation(s)
- YanDan Du
- Department of Clinical Laboratory, Inner Mongolia Forestry General Hospital (The second Clinical Medical School of Inner Mongolia, University for the Nationalities), Hulunbuir, Inner Mongolia, China
| | - ZhiHui Mi
- Inner Mongolia Di An Feng Xin Medical Technology Co., LTD, Huhhot, Inner Mongolia, China
| | - YaPing Xie
- SCIEX China Technology Co., Beijing, China
| | - DeSheng Lu
- Department of Clinical Laboratory, Inner Mongolia Forestry General Hospital (The second Clinical Medical School of Inner Mongolia, University for the Nationalities), Hulunbuir, Inner Mongolia, China
| | - HaiJun Zheng
- Department of Clinical Laboratory, Inner Mongolia Forestry General Hospital (The second Clinical Medical School of Inner Mongolia, University for the Nationalities), Hulunbuir, Inner Mongolia, China
| | - Hui Sun
- Department of Clinical Laboratory, Inner Mongolia Forestry General Hospital (The second Clinical Medical School of Inner Mongolia, University for the Nationalities), Hulunbuir, Inner Mongolia, China
| | - Meng Zhang
- Inner Mongolia Di An Feng Xin Medical Technology Co., LTD, Huhhot, Inner Mongolia, China
- * E-mail: (MZ); (YQN)
| | - YiQing Niu
- Department of Clinical Laboratory, Inner Mongolia Forestry General Hospital (The second Clinical Medical School of Inner Mongolia, University for the Nationalities), Hulunbuir, Inner Mongolia, China
- * E-mail: (MZ); (YQN)
| |
Collapse
|
38
|
Kim TK, Tirloni L, Bencosme-Cuevas E, Kim TH, Diedrich JK, Yates JR, Mulenga A. Borrelia burgdorferi infection modifies protein content in saliva of Ixodes scapularis nymphs. BMC Genomics 2021; 22:152. [PMID: 33663385 PMCID: PMC7930271 DOI: 10.1186/s12864-021-07429-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Lyme disease (LD) caused by Borrelia burgdorferi is the most prevalent tick-borne disease. There is evidence that vaccines based on tick proteins that promote tick transmission of B. burgdorferi could prevent LD. As Ixodes scapularis nymph tick bites are responsible for most LD cases, this study sought to identify nymph tick saliva proteins associated with B. burgdorferi transmission using LC-MS/MS. Tick saliva was collected using a non-invasive method of stimulating ticks (uninfected and infected: unfed, and every 12 h during feeding through 72 h, and fully-fed) to salivate into 2% pilocarpine-PBS for protein identification using LC-MS/MS. RESULTS We identified a combined 747 tick saliva proteins of uninfected and B. burgdorferi infected ticks that were classified into 25 functional categories: housekeeping-like (48%), unknown function (18%), protease inhibitors (9%), immune-related (6%), proteases (8%), extracellular matrix (7%), and small categories that account for <5% each. Notably, B. burgdorferi infected ticks secreted high number of saliva proteins (n=645) than uninfected ticks (n=376). Counter-intuitively, antimicrobial peptides, which function to block bacterial infection at tick feeding site were suppressed 23-85 folds in B. burgdorferi infected ticks. Similar to glycolysis enzymes being enhanced in mammalian cells exposed to B. burgdorferi : eight of the 10-glycolysis pathway enzymes were secreted at high abundance by B. burgdorferi infected ticks. Of significance, rabbits exposed to B. burgdorferi infected ticks acquired potent immunity that caused 40-60% mortality of B. burgdorferi infected ticks during the second infestation compared to 15-28% for the uninfected. This might be explained by ELISA data that show that high expression levels of immunogenic proteins in B. burgdorferi infected ticks. CONCLUSION Data here suggest that B. burgdorferi infection modified protein content in tick saliva to promote its survival at the tick feeding site. For instance, enzymes; copper/zinc superoxide dismutase that led to production of H2O2 that is toxic to B. burgdorferi were suppressed, while, catalase and thioredoxin that neutralize H2O2, and pyruvate kinase which yields pyruvate that protects Bb from H2O2 killing were enhanced. We conclude data here is an important resource for discovery of effective antigens for a vaccine to prevent LD.
Collapse
Affiliation(s)
- Tae Kwon Kim
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
- Department of Diagnostic Medicine and Veterinary Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, United States of America
| | - Lucas Tirloni
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
- Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, Hamilton, Montana, United States of America
| | - Emily Bencosme-Cuevas
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
| | - Tae Heung Kim
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
| | - Jolene K Diedrich
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
- Mass Spectrometry Core, Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - John R Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Albert Mulenga
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America.
| |
Collapse
|
39
|
Boulanger N, Wikel S. Induced Transient Immune Tolerance in Ticks and Vertebrate Host: A Keystone of Tick-Borne Diseases? Front Immunol 2021; 12:625993. [PMID: 33643313 PMCID: PMC7907174 DOI: 10.3389/fimmu.2021.625993] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/22/2021] [Indexed: 12/23/2022] Open
Abstract
Ticks and tick transmitted infectious agents are increasing global public health threats due to increasing abundance, expanding geographic ranges of vectors and pathogens, and emerging tick-borne infectious agents. Greater understanding of tick, host, and pathogen interactions will contribute to development of novel tick control and disease prevention strategies. Tick-borne pathogens adapt in multiple ways to very different tick and vertebrate host environments and defenses. Ticks effectively pharmacomodulate by its saliva host innate and adaptive immune defenses. In this review, we examine the idea that successful synergy between tick and tick-borne pathogen results in host immune tolerance that facilitates successful tick infection and feeding, creates a favorable site for pathogen introduction, modulates cutaneous and systemic immune defenses to establish infection, and contributes to successful long-term infection. Tick, host, and pathogen elements examined here include interaction of tick innate immunity and microbiome with tick-borne pathogens; tick modulation of host cutaneous defenses prior to pathogen transmission; how tick and pathogen target vertebrate host defenses that lead to different modes of interaction and host infection status (reservoir, incompetent, resistant, clinically ill); tick saliva bioactive molecules as important factors in determining those pathogens for which the tick is a competent vector; and, the need for translational studies to advance this field of study. Gaps in our understanding of these relationships are identified, that if successfully addressed, can advance the development of strategies to successfully disrupt both tick feeding and pathogen transmission.
Collapse
Affiliation(s)
- Nathalie Boulanger
- Fédération de Médecine Translationnelle - UR7290, Early Bacterial Virulence, Group Borrelia, Université de Strasbourg, Strasbourg, France.,Centre National de Référence Borrelia, Centre Hospitalier Universitaire, Strasbourg, France
| | - Stephen Wikel
- Department of Medical Sciences, Frank H. Netter, M.D., School of Medicine, Quinnipiac University, Hamden, CT, United States
| |
Collapse
|
40
|
Mateos-Hernández L, Pipová N, Allain E, Henry C, Rouxel C, Lagrée AC, Haddad N, Boulouis HJ, Valdés JJ, Alberdi P, de la Fuente J, Cabezas-Cruz A, Šimo L. Enlisting the Ixodes scapularis Embryonic ISE6 Cell Line to Investigate the Neuronal Basis of Tick-Pathogen Interactions. Pathogens 2021; 10:pathogens10010070. [PMID: 33466622 PMCID: PMC7828734 DOI: 10.3390/pathogens10010070] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 01/03/2023] Open
Abstract
Neuropeptides are small signaling molecules expressed in the tick central nervous system, i.e., the synganglion. The neuronal-like Ixodes scapularis embryonic cell line, ISE6, is an effective tool frequently used for examining tick–pathogen interactions. We detected 37 neuropeptide transcripts in the I. scapularis ISE6 cell line using in silico methods, and six of these neuropeptide genes were used for experimental validation. Among these six neuropeptide genes, the tachykinin-related peptide (TRP) of ISE6 cells varied in transcript expression depending on the infection strain of the tick-borne pathogen, Anaplasma phagocytophilum. The immunocytochemistry of TRP revealed cytoplasmic expression in a prominent ISE6 cell subpopulation. The presence of TRP was also confirmed in A. phagocytophilum-infected ISE6 cells. The in situ hybridization and immunohistochemistry of TRP of I. scapularis synganglion revealed expression in distinct neuronal cells. In addition, TRP immunoreaction was detected in axons exiting the synganglion via peripheral nerves as well as in hemal nerve-associated lateral segmental organs. The characterization of a complete Ixodes neuropeptidome in ISE6 cells may serve as an effective in vitro tool to study how tick-borne pathogens interact with synganglion components that are vital to tick physiology. Therefore, our current study is a potential stepping stone for in vivo experiments to further examine the neuronal basis of tick–pathogen interactions.
Collapse
Affiliation(s)
- Lourdes Mateos-Hernández
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (L.M.-H.); (E.A.); (C.R.); (A.-C.L.); (N.H.); (H.-J.B.)
| | - Natália Pipová
- Faculty of Science, Pavol Jozef Šafarik University in Košice, 04180 Košice, Slovakia;
| | - Eléonore Allain
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (L.M.-H.); (E.A.); (C.R.); (A.-C.L.); (N.H.); (H.-J.B.)
| | - Céline Henry
- AgroParisTech, Micalis Institute, Université Paris-Saclay, PAPPSO, INRAE, 78350 Jouy-en-Josas, France;
| | - Clotilde Rouxel
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (L.M.-H.); (E.A.); (C.R.); (A.-C.L.); (N.H.); (H.-J.B.)
| | - Anne-Claire Lagrée
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (L.M.-H.); (E.A.); (C.R.); (A.-C.L.); (N.H.); (H.-J.B.)
| | - Nadia Haddad
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (L.M.-H.); (E.A.); (C.R.); (A.-C.L.); (N.H.); (H.-J.B.)
| | - Henri-Jean Boulouis
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (L.M.-H.); (E.A.); (C.R.); (A.-C.L.); (N.H.); (H.-J.B.)
| | - James J. Valdés
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, 37005 Ceske Budejovice, Czech Republic;
- Department of Virology, Veterinary Research Institute, Hudcova 70, 62100 Brno, Czech Republic
| | - Pilar Alberdi
- SaBio Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (P.A.); (J.d.l.F.)
- Neuroplasticity and Neurodegeneration Group, Regional Centre for Biomedical Research (CRIB), Ciu-dad Real Medical School, University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - José de la Fuente
- SaBio Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (P.A.); (J.d.l.F.)
- Center for Veterinary Health Sciences, Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078, USA
| | - Alejandro Cabezas-Cruz
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (L.M.-H.); (E.A.); (C.R.); (A.-C.L.); (N.H.); (H.-J.B.)
- Correspondence: (A.C.-C.); (L.Š.); Tel.: +33-6-31-23-51-91 (A.C.-C.); +33-1-49-77-46-52 (L.Š.)
| | - Ladislav Šimo
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d’Alfort, Paris-Est Sup, 94700 Maisons-Alfort, France; (L.M.-H.); (E.A.); (C.R.); (A.-C.L.); (N.H.); (H.-J.B.)
- Correspondence: (A.C.-C.); (L.Š.); Tel.: +33-6-31-23-51-91 (A.C.-C.); +33-1-49-77-46-52 (L.Š.)
| |
Collapse
|
41
|
Martins LA, Palmisano G, Cortez M, Kawahara R, de Freitas Balanco JM, Fujita A, Alonso BI, Barros-Battesti DM, Braz GRC, Tirloni L, Esteves E, Daffre S, Fogaça AC. The intracellular bacterium Rickettsia rickettsii exerts an inhibitory effect on the apoptosis of tick cells. Parasit Vectors 2020; 13:603. [PMID: 33261663 PMCID: PMC7706286 DOI: 10.1186/s13071-020-04477-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Rickettsia rickettsii is a tick-borne obligate intracellular bacterium that causes Rocky Mountain spotted fever, a life-threatening illness. To obtain an insight into the vector-pathogen interactions, we assessed the effects of infection with R. rickettsii on the proteome cells of the tick embryonic cell line BME26. METHODS The proteome of BME26 cells was determined by label-free high-performance liquid chromatography coupled with tandem mass spectrometry analysis. Also evaluated were the effects of infection on the activity of caspase-3, assessed by the hydrolysis of a synthetic fluorogenic substrate in enzymatic assays, and on the exposition of phosphatidyserine, evaluated by live-cell fluorescence microscopy after labeling with annexin-V. Finally, the effects of activation or inhibition of caspase-3 activity on the growth of R. rickettsii in BME26 cells was determined. RESULTS Tick proteins of different functional classes were modulated in a time-dependent manner by R. rickettsii infection. Regarding proteins involved in apoptosis, certain negative regulators were downregulated at the initial phase of the infection (6 h) but upregulated in the middle of the exponential phase of the bacterial growth (48 h). Microorganisms are known to be able to inhibit apoptosis of the host cell to ensure their survival and proliferation. We therefore evaluated the effects of infection on classic features of apoptotic cells and observed DNA fragmentation exclusively in noninfected cells. Moreover, both caspase-3 activity and phosphatidylserine exposition were lower in infected than in noninfected cells. Importantly, while the activation of caspase-3 exerted a detrimental effect on rickettsial proliferation, its inhibition increased bacterial growth. CONCLUSIONS Taken together, these results show that R. rickettsii modulates the proteome and exerts an inhibitory effect on apoptosis in tick cellsthat seems to be important to ensure cell colonization.
Collapse
Affiliation(s)
- Larissa Almeida Martins
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.,Rocky Mountain Laboratories, National Institutes of Health, National Institute of Allergy and Infectious Diseases, Hamilton, USA
| | - Giuseppe Palmisano
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Mauro Cortez
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Rebeca Kawahara
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.,Department of Molecular Sciences, Macquarie University, Sydney, NSW, Australia
| | | | - André Fujita
- Department of Computational Science, Institute of Mathematics and Statistics, University of São Paulo, São Paulo, SP, Brazil
| | - Beatriz Iglesias Alonso
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | | | - Gloria Regina Cardoso Braz
- Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Lucas Tirloni
- Rocky Mountain Laboratories, National Institutes of Health, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA
| | - Eliane Esteves
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Sirlei Daffre
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Andréa Cristina Fogaça
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
42
|
Xi B, Luo J, Gao YQ, Yang XL, Guo TF, Li WH, Du TQ. Transcriptome-metabolome analysis of fatty acid of Bamei pork and Gansu Black pork in China. Bioprocess Biosyst Eng 2020; 44:995-1002. [PMID: 33159544 DOI: 10.1007/s00449-020-02468-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 10/22/2020] [Indexed: 01/23/2023]
Abstract
To study the difference in transcriptome level of fatty acid metabolism pathway in Bamei pork and the difference of pork quality caused by the difference. In this study, Bamei pigs breeding in Huzhu farm of QingHai province were selected as the test object, compared with Gansu Black pigs. Four indexes of nutmeg acid (DX1), palmitic acid (DX2), stearic acid (DX3) and linoleic acid (DX4) were set. The expression profiles of fat metabolism related genes between the two groups samples were analysed by GCMS metabolomics and transcriptomics, then coexpression network analysis were conducted to obtain phenotypic related genes. The results showed that the metabolic levels of DX3 and DX4 were significantly higher than those of other fatty acids. Among these differences, the ENSSSCG00000024681 (G1) and ENSSSCG00000036883 (G2) genes play important regulatory roles in fatty acid metabolism, and the upregulated expression of their gene obviously affects the level of fatty acid metabolism, thereby affecting the quality and taste of pork. In addition, we found that there was a good correlation between the same lines, and the genetic traits of the hybrid lines of Bamei pig and Black pig are more inclined to Bamei pig. In the independent fatty acid metabolism, "Mg2+"and flavin adenine dinucleotide are more active, which plays an important role in energy utilization. Therefore, we can be inferred that the metabolism of stearic acid and linoleic acid are important fatty acids for pork quality. It also further confirms that the research method of combined omics is of great significance for the study of species traits and gene functions.
Collapse
Affiliation(s)
- Bin Xi
- Lanzhou Institute of Animal Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, Gansu, China.,Laboratory of Quality and Safety Risk Assessment for Livestock Products (Lanzhou), Ministry of Agriculture, Lanzhou, 730050, Gansu, China
| | - Jin Luo
- Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, Gansu, China
| | - Ya-Qin Gao
- Lanzhou Institute of Animal Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, Gansu, China. .,Laboratory of Quality and Safety Risk Assessment for Livestock Products (Lanzhou), Ministry of Agriculture, Lanzhou, 730050, Gansu, China.
| | - Xiao-Ling Yang
- Lanzhou Institute of Animal Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, Gansu, China.,Laboratory of Quality and Safety Risk Assessment for Livestock Products (Lanzhou), Ministry of Agriculture, Lanzhou, 730050, Gansu, China
| | - Tian-Fen Guo
- Lanzhou Institute of Animal Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, Gansu, China.,Laboratory of Quality and Safety Risk Assessment for Livestock Products (Lanzhou), Ministry of Agriculture, Lanzhou, 730050, Gansu, China
| | - Wei-Hong Li
- Lanzhou Institute of Animal Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, Gansu, China.,Laboratory of Quality and Safety Risk Assessment for Livestock Products (Lanzhou), Ministry of Agriculture, Lanzhou, 730050, Gansu, China
| | - Tian-Qing Du
- Lanzhou Institute of Animal Husbandry and Pharmaceutical Science, Chinese Academy of Agricultural Sciences, Lanzhou, 730050, Gansu, China.,Laboratory of Quality and Safety Risk Assessment for Livestock Products (Lanzhou), Ministry of Agriculture, Lanzhou, 730050, Gansu, China
| |
Collapse
|
43
|
Mans BJ. Quantitative Visions of Reality at the Tick-Host Interface: Biochemistry, Genomics, Proteomics, and Transcriptomics as Measures of Complete Inventories of the Tick Sialoverse. Front Cell Infect Microbiol 2020; 10:574405. [PMID: 33042874 PMCID: PMC7517725 DOI: 10.3389/fcimb.2020.574405] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/13/2020] [Indexed: 12/19/2022] Open
Abstract
Species have definitive genomes. Even so, the transcriptional and translational products of the genome are dynamic and subject to change over time. This is especially true for the proteins secreted by ticks at the tick-host feeding interface that represent a complex system known as the sialoverse. The sialoverse represent all of the proteins derived from tick salivary glands for all tick species that may be involved in tick-host interaction and the modulation of the host's defense mechanisms. The current study contemplates the advances made over time to understand and describe the complexity present in the sialoverse. Technological advances at given periods in time allowed detection of functions, genes, and proteins enabling a deeper insight into the complexity of the sialoverse and a concomitant expansion in complexity with as yet, no end in sight. The importance of systematic classification of the sialoverse is highlighted with the realization that our coverage of transcriptome and proteome space remains incomplete, but that complete descriptions may be possible in the future. Even so, analysis and integration of the sialoverse into a comprehensive understanding of tick-host interactions may require further technological advances given the high level of expected complexity that remains to be uncovered.
Collapse
Affiliation(s)
- Ben J Mans
- Epidemiology, Parasites and Vectors, Agricultural Research Council-Onderstepoort Veterinary Research, Pretoria, South Africa.,Department of Veterinary Tropical Diseases, University of Pretoria, Pretoria, South Africa.,Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
| |
Collapse
|
44
|
Ndawula C, Tabor AE. Cocktail Anti-Tick Vaccines: The Unforeseen Constraints and Approaches toward Enhanced Efficacies. Vaccines (Basel) 2020; 8:E457. [PMID: 32824962 PMCID: PMC7564958 DOI: 10.3390/vaccines8030457] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 12/17/2022] Open
Abstract
Ticks are second to mosquitoes as vectors of disease. Ticks affect livestock industries in Asia, Africa and Australia at ~$1.13 billion USD per annum. For instance, 80% of the global cattle population is at risk of infestation by the Rhipicephalus microplus species-complex, which in 2016 was estimated to cause $22-30 billion USD annual losses. Although the management of tick populations mainly relies on the application of acaricides, this raises concerns due to tick resistance and accumulation of chemical residues in milk, meat, and the environment. To counteract acaricide-resistant tick populations, immunological tick control is regarded among the most promising sustainable strategies. Indeed, immense efforts have been devoted toward identifying tick vaccine antigens. Until now, Bm86-based vaccines have been the most effective under field conditions, but they have shown mixed success worldwide. Currently, of the two Bm86 vaccines commercialized in the 1990s (GavacTM in Cuba and TickGARDPLUSTM in Australia), only GavacTM is available. There is thus growing consensus that combining antigens could broaden the protection range and enhance the efficacies of tick vaccines. Yet, the anticipated outcomes have not been achieved under field conditions. Therefore, this review demystifies the potential limitations and proposes ways of sustaining enhanced cocktail tick vaccine efficacy.
Collapse
Affiliation(s)
- Charles Ndawula
- Vaccinology Research program, National Livestock Resources Research Institute, P O. Box 5746, Nakyesasa 256, Uganda
| | - Ala E. Tabor
- Centre for Animal Science, Queensland Alliance for Agriculture & Food Innovation, The University of Queensland Australia, St Lucia 4072, Queensland, Australia
- School of Chemistry & Molecular Biosciences, The University of Queensland, St Lucia 4072, Queensland, Australia
| |
Collapse
|
45
|
Benelli G. Pathogens Manipulating Tick Behavior-Through a Glass, Darkly. Pathogens 2020; 9:pathogens9080664. [PMID: 32824571 PMCID: PMC7459789 DOI: 10.3390/pathogens9080664] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/05/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022] Open
Abstract
Pathogens can manipulate the phenotypic traits of their hosts and vectors, maximizing their own fitness. Among the phenotypic traits that can be modified, manipulating vector behavior represents one of the most fascinating facets. How pathogens infection affects behavioral traits of key insect vectors has been extensively investigated. Major examples include Plasmodium, Leishmania and Trypanosoma spp. manipulating the behavior of mosquitoes, sand flies and kissing bugs, respectively. However, research on how pathogens can modify tick behavior is patchy. This review focuses on current knowledge about the behavioral changes triggered by Anaplasma, Borrelia, Babesia, Bartonella, Rickettsia and tick-borne encephalitis virus (TBEV) infection in tick vectors, analyzing their potential adaptive significance. As a general trend, being infected by Borrelia and TBEV boosts tick mobility (both questing and walking activity). Borrelia and Anaplasma infection magnifies Ixodes desiccation resistance, triggering physiological changes (Borrelia: higher fat reserves; Anaplasma: synthesis of heat shock proteins). Anaplasma infection also improves cold resistance in infected ticks through synthesis of an antifreeze glycoprotein. Being infected by Anaplasma, Borrelia and Babesia leads to increased tick survival. Borrelia, Babesia and Bartonella infection facilitates blood engorgement. In the last section, current challenges for future studies are outlined.
Collapse
Affiliation(s)
- Giovanni Benelli
- Department of Agriculture, Food and Environment, University of Pisa, via del Borghetto 80, 56124 Pisa, Italy
| |
Collapse
|
46
|
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: 10] [Impact Index Per Article: 2.0] [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
|
47
|
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: 11] [Impact Index Per Article: 2.2] [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
|
48
|
Zheng W, Umemiya-Shirafuji R, Chen S, Okado K, Adjou Moumouni PF, Suzuki H, Yang S, Liu M, Xuan X. Identification of Haemaphysalis longicornis Genes Differentially Expressed in Response to Babesia microti Infection. Pathogens 2020; 9:E378. [PMID: 32423088 PMCID: PMC7281432 DOI: 10.3390/pathogens9050378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/02/2020] [Accepted: 05/12/2020] [Indexed: 12/31/2022] Open
Abstract
Haemaphysalis longicornis is a tick and a vector of various pathogens, including the human pathogenetic Babesia microti. The objective of this study was to identify female H. longicornis genes differentially expressed in response to infection with B. microti Gray strain by using a suppression subtractive hybridization (SSH) procedure. A total of 302 randomly selected clones were sequenced and analyzed in the forward subtracted SSH cDNA library related to Babesia infection, and 110 clones in the reverse cDNA library. Gene ontology assignments and sequence analyses of tick sequences in the forward cDNA library showed that 14 genes were related to response to stimulus or/and immune system process, and 7 genes had the higher number of standardized sequences per kilobase (SPK). Subsequent real-time PCR detection showed that eight genes including those encoding for Obg-like ATPase 1 (ola1), Calreticulin (crt), vitellogenin 1 (Vg1) and Vg2 were up-regulated in fed ticks. Compared to uninfected ticks, infected ticks had six up-regulated genes, including ola1, crt and Vg2. Functional analysis of up-regulated genes in fed or Babesia-infected ticks by RNA interference showed that knockdown of crt and Vg2 in infected ticks and knockdown of ola1 in uninfected ticks accelerated engorgement. In contrast, Vg1 knockdown in infected ticks had delayed engorgement. Knockdown of crt and Vg1 in infected ticks decreased engorged female weight. Vg2 knockdown reduced B. microti infection levels by 51% when compared with controls. The results reported here increase our understanding of roles of H. longicornis genes in blood feeding and B. microti infection.
Collapse
Affiliation(s)
- Weiqing Zheng
- The Collaboration Unit for Field Epidemiology of State Key Laboratory for Infectious Disease Prevention and Control, Jiangxi Provincial key Laboratory of Animal-Origin and Vector-Borne Diseases, Nanchang Center for Disease Control and Prevention, Honggutan New District, Nanchang 330038, China; (W.Z.); (S.C.)
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido 080-8555, Japan; (K.O.); (P.F.A.M.); (H.S.); (M.L.)
| | - Rika Umemiya-Shirafuji
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido 080-8555, Japan; (K.O.); (P.F.A.M.); (H.S.); (M.L.)
| | - Shengen Chen
- The Collaboration Unit for Field Epidemiology of State Key Laboratory for Infectious Disease Prevention and Control, Jiangxi Provincial key Laboratory of Animal-Origin and Vector-Borne Diseases, Nanchang Center for Disease Control and Prevention, Honggutan New District, Nanchang 330038, China; (W.Z.); (S.C.)
| | - Kiyoshi Okado
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido 080-8555, Japan; (K.O.); (P.F.A.M.); (H.S.); (M.L.)
| | - Paul Franck Adjou Moumouni
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido 080-8555, Japan; (K.O.); (P.F.A.M.); (H.S.); (M.L.)
| | - Hiroshi Suzuki
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido 080-8555, Japan; (K.O.); (P.F.A.M.); (H.S.); (M.L.)
| | - Shu Yang
- The Collaboration Unit for Field Epidemiology of State Key Laboratory for Infectious Disease Prevention and Control, Jiangxi Provincial key Laboratory of Animal-Origin and Vector-Borne Diseases, Nanchang Center for Disease Control and Prevention, Honggutan New District, Nanchang 330038, China; (W.Z.); (S.C.)
| | - Mingming Liu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido 080-8555, Japan; (K.O.); (P.F.A.M.); (H.S.); (M.L.)
| | - Xuenan Xuan
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido 080-8555, Japan; (K.O.); (P.F.A.M.); (H.S.); (M.L.)
| |
Collapse
|
49
|
Villar M, Pacheco I, Merino O, Contreras M, Mateos-Hernández L, Prado E, Barros-Picanço DK, Lima-Barbero JF, Artigas-Jerónimo S, Alberdi P, Fernández de Mera IG, Estrada-Peña A, Cabezas-Cruz A, de la Fuente J. Tick and Host Derived Compounds Detected in the Cement Complex Substance. Biomolecules 2020; 10:E555. [PMID: 32260542 PMCID: PMC7226240 DOI: 10.3390/biom10040555] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 02/07/2023] Open
Abstract
Ticks are obligate hematophagous arthropods and vectors of pathogens affecting human and animal health worldwide. Cement is a complex protein polymerization substance secreted by ticks with antimicrobial properties and a possible role in host attachment, sealing the feeding lesion, facilitating feeding and pathogen transmission, and protection from host immune and inflammatory responses. The biochemical properties of tick cement during feeding have not been fully characterized. In this study, we characterized the proteome of Rhipicephalus microplus salivary glands (sialome) and cement (cementome) together with their physicochemical properties at different adult female parasitic stages. The results showed the combination of tick and host derived proteins and other biomolecules such as α-Gal in cement composition, which varied during the feeding process. We propose that these compounds may synergize in cement formation, solidification and maintenance to facilitate attachment, feeding, interference with host immune response and detachment. These results advanced our knowledge of the complex tick cement composition and suggested that tick and host derived compounds modulate cement properties throughout tick feeding.
Collapse
Affiliation(s)
- Margarita Villar
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
- Biochemistry Section, Faculty of Science and Chemical Technologies, and Regional Centre for Biomedical Research (CRIB), University of Castilla-La Mancha, 13071 Ciudad Real, Spain
| | - Iván Pacheco
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
| | - Octavio Merino
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Tamaulipas, Km 5, Carretera Victoria-Mante, CP 87000 Ciudad Victoria, Tamaulipas, Mexico;
| | - Marinela Contreras
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
| | - Lourdes Mateos-Hernández
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
- UMR BIPAR, INRAE, ANSES, Ecole Nationale Vétérinaire d’Alfort, Université Paris-Est, 94700 Maisons-Alfort, France;
| | - Eduardo Prado
- Department of Applied Physics, Faculty of Chemical Sciences and Technologies, Universidad de Castilla-La Mancha, Avda. Camilo José Cela 10, 13071 Ciudad Real, Spain;
| | - Dina Karen Barros-Picanço
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
| | - José Francisco Lima-Barbero
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
- Sabiotec, Camino de Moledores s/n. 13003, 13071 Ciudad Real, Spain
| | - Sara Artigas-Jerónimo
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
| | - Pilar Alberdi
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
| | - Isabel G. Fernández de Mera
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
| | | | - Alejandro Cabezas-Cruz
- UMR BIPAR, INRAE, ANSES, Ecole Nationale Vétérinaire d’Alfort, Université Paris-Est, 94700 Maisons-Alfort, France;
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC-CSIC-UCLM-JCCM), Ronda de Toledo s/n, 13005 Ciudad Real, Spain; (M.V.); (I.P.); (M.C.); (L.M.-H.); (D.K.B.-P.); (J.F.L.-B.); (S.A.-J.); (P.A.); (I.G.F.d.M.)
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
50
|
Pathological Mechanistic Studies of Osimertinib Resistance in Non-Small-Cell Lung Cancer Cells Using an Integrative Metabolomics-Proteomics Analysis. JOURNAL OF ONCOLOGY 2020; 2020:6249829. [PMID: 32256584 PMCID: PMC7103047 DOI: 10.1155/2020/6249829] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/14/2020] [Accepted: 01/18/2020] [Indexed: 12/14/2022]
Abstract
Background Osimertinib is the first-line therapeutic option for the T790M-mutant non-small-cell lung cancer and the acquired resistance obstructs its application. It is an urgent challenge to identify the potential mechanisms of osimertinib resistance for uncovering some novel therapeutic approaches. Methods In the current study, the cell metabolomics based on ultra-high-performance liquid chromatography coupled with linear ion trap-Orbitrap mass spectrometry and the qualitative and tandem mass tags quantitative proteomics were performed. Results 54 differential metabolites and 195 differentially expressed proteins were, respectively, identified. The amino acids metabolisms were significantly altered. HIF-1 signaling pathway modulating P-glycoproteins expression, PI3K-Akt pathway regulating survivin expression, and oxidative phosphorylation were upregulated, while arginine and proline metabolism regulating NO production and glycolysis/gluconeogenesis were downregulated during osimertinib resistance. Conclusion The regulation of HIF-1 and PI3K-Akt signaling pathway, energy supply process, and amino acids metabolism are the promising therapeutic tactics for osimertinib resistance.
Collapse
|