1
|
Cecilio P, Rogerio LA, D Serafim T, Tang K, Willen L, Iniguez E, Meneses C, Chaves LF, Zhang Y, Dos Santos Felix L, Huang W, Garcia Guizzo M, Castañeda-Casado P, Jacobs-Lorena M, Valenzuela JG, Rodrigues J, Oliveira F. Leishmania sand fly-transmission is disrupted by Delftia tsuruhatensis TC1 bacteria. Nat Commun 2025; 16:3571. [PMID: 40341020 PMCID: PMC12062286 DOI: 10.1038/s41467-025-58769-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 04/02/2025] [Indexed: 05/10/2025] Open
Abstract
Most human pathogenic Leishmania species are zoonotic agents; therefore, sand fly-based control strategies are essential to prevent parasite circulation. Here, we used the Delftia tsuruhatensis TC1 strain, that inhibits the development of Plasmodium in mosquitoes, but in the context of Leishmania-infected sand flies. We show that D. tsuruhatensis TC1 colonizes the midgut of Phlebotomus duboscqi sand flies and impacts the development of L. major parasites, independently of the colonization timing. This phenotype is likely an indirect consequence of D. tsuruhatensis colonization, related with the induction of sand fly gut dysbiosis. Importantly, Leishmania-infected, D. tsuruhatensis-fed sand flies are less able to transmit L. major parasites and cause disease in mice. Modelling supports the disruption of disease endemicity in the field, highlighting D. tsuruhatensis as a promising agent for the control of leishmaniasis.
Collapse
Affiliation(s)
- Pedro Cecilio
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| | - Luana A Rogerio
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National; Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Tiago D Serafim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National; Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Kristina Tang
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National; Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Laura Willen
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National; Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Eva Iniguez
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National; Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Claudio Meneses
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National; Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Luis F Chaves
- Department of Environmental and Occupational Health, School of Public Health-Bloomington, and Department of Geography, Indiana University, Bloomington, IN, USA
| | - Yue Zhang
- Integrated Data Sciences Section (IDSS), Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Luiza Dos Santos Felix
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National; Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Wei Huang
- Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Melina Garcia Guizzo
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | | | - Marcelo Jacobs-Lorena
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National; Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | | | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National; Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, Phnom Penh, Cambodia.
| |
Collapse
|
2
|
Hodžić A, Duscher GG, Alić A, Beck R, Berry D. Peritrophic matrix: an important determinant of vector competence in hematophagous arthropods. Trends Parasitol 2025; 41:374-386. [PMID: 40148178 DOI: 10.1016/j.pt.2025.03.005] [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: 01/24/2025] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025]
Abstract
The peritrophic matrix (PM) is a non-cellular, glycan-rich structure that lines the gut epithelium of most invertebrates, including arthropod vectors that transmit diseases of public health and veterinary concern. This semipermeable barrier, functionally analogous to the vertebrate mucosal layer, separates the gut lumen from epithelial cells and provides protection against invading pathogens and their toxins. Beyond its mechanical protective role in the gut, the PM plays a crucial part in arthropod innate immunity. Here, we summarize the most recent advances in understanding the molecular mechanisms of vector-pathogen interactions in blood-feeding arthropods and discuss the significance of the PM in modulating vector competence. This knowledge could contribute to the development of novel strategies to control vector-borne infections.
Collapse
Affiliation(s)
- Adnan Hodžić
- Centre for Microbiology and Environmental Systems Science (CMESS), Department of Microbiology and Ecosystem Science, Division of Microbial Ecology (DoME), University of Vienna, 1030 Vienna, Austria.
| | - Georg Gerhard Duscher
- AGES Research Services, Austrian Agency for Health and Food Safety, 2340 Mödling, Austria
| | - Amer Alić
- Department of Clinical Sciences of Veterinary Medicine, Faculty of Veterinary Medicine, University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina
| | - Relja Beck
- Department for Bacteriology and Parasitology, Croatian Veterinary Institute, 10000 Zagreb, Croatia
| | - David Berry
- Centre for Microbiology and Environmental Systems Science (CMESS), Department of Microbiology and Ecosystem Science, Division of Microbial Ecology (DoME), University of Vienna, 1030 Vienna, Austria; Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, 1030 Vienna, Austria
| |
Collapse
|
3
|
Liu Y, Dong X, Sun L, Cui H, Kang J, Bu N, Zhang Y, Qi Z, Li Z, Zhang Z, Zhao L. Analysis of the Microbial Community Structure of Ixodes persulcatus at Each Developmental Stage. Animals (Basel) 2025; 15:830. [PMID: 40150359 PMCID: PMC11939619 DOI: 10.3390/ani15060830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Ticks are the second most significant vector of pathogens worldwide. Ixodes persulcatus is one of the dominant tick species in Inner Mongolia that can carry and transmit various pathogenic microorganisms. However, only one specific pathogen has been detected in a particular developmental stage of I. persulcatus, moreover metagenomic analysis has been conducted only in the adult tick stage. In this study, we used I. persulcatus at different developmental stages (first-generation female adult ticks, eggs, larval ticks, engorged larval ticks, nymphal ticks, engorged nymphal ticks, and second-generation adult ticks) from Inner Mongolia as materials for nucleic acid extraction. Subsequently, we constructed Illumina PE250 and Illumina PE150 libraries and sequenced them on the Illumina NovaSeq 6000 platform. Finally, we used molecular biology software and sequence analysis platform to analyze microbial community structures. Illumina PE250 sequencing revealed that the seven developmental stages of I. persulcatus were annotated to 21 phyla, 43 classes, 104 orders, 188 families, 391 genera, and 556 species of bacteria. Among them, 4 phyla and 14 genera were present at all developmental stages, with Proteobacteria being the dominant phylum and Rickettsia spp. being the dominant genus. In addition, Rickettsia had the highest relative abundance in the seven developmental stages. All developmental stages were annotated to a certain abundance of Brucella spp. Illumina PE150 sequencing revealed that the three samples (X-I-YDCP: first-generation adult ticks; X-I-MIX: mixed samples of eggs, larval ticks, and nymphal ticks; X-I-EDCP: second-generation adult ticks) of I. persulcatus were annotated to six orders, 28 families, 72 genera, and 158 species of viruses, of which 46 genera and 80 species were found in all three sample species. To the best of our knowledge, this is the first study that comprehensively analyzed the microbial community composition of I. persulcatus at different developmental stages. Based on the study outcomes, certain abundance of Rickettsia japonica, bovine viral diarrhea virus, and African swine fever virus were annotated to I. persulcatus.
Collapse
Affiliation(s)
- Yonghong Liu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.L.); (X.D.); (L.S.); (H.C.); (J.K.); (N.B.); (Y.Z.); (Z.Q.); (Z.L.); (Z.Z.)
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture and Rural Affairs, Hohhot 010010, China
| | - Xiaonan Dong
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.L.); (X.D.); (L.S.); (H.C.); (J.K.); (N.B.); (Y.Z.); (Z.Q.); (Z.L.); (Z.Z.)
| | - Lianyang Sun
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.L.); (X.D.); (L.S.); (H.C.); (J.K.); (N.B.); (Y.Z.); (Z.Q.); (Z.L.); (Z.Z.)
| | - Hao Cui
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.L.); (X.D.); (L.S.); (H.C.); (J.K.); (N.B.); (Y.Z.); (Z.Q.); (Z.L.); (Z.Z.)
| | - Jiamei Kang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.L.); (X.D.); (L.S.); (H.C.); (J.K.); (N.B.); (Y.Z.); (Z.Q.); (Z.L.); (Z.Z.)
| | - Nan Bu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.L.); (X.D.); (L.S.); (H.C.); (J.K.); (N.B.); (Y.Z.); (Z.Q.); (Z.L.); (Z.Z.)
| | - Yishuai Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.L.); (X.D.); (L.S.); (H.C.); (J.K.); (N.B.); (Y.Z.); (Z.Q.); (Z.L.); (Z.Z.)
| | - Zehao Qi
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.L.); (X.D.); (L.S.); (H.C.); (J.K.); (N.B.); (Y.Z.); (Z.Q.); (Z.L.); (Z.Z.)
| | - Zixuan Li
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.L.); (X.D.); (L.S.); (H.C.); (J.K.); (N.B.); (Y.Z.); (Z.Q.); (Z.L.); (Z.Z.)
| | - Zilong Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.L.); (X.D.); (L.S.); (H.C.); (J.K.); (N.B.); (Y.Z.); (Z.Q.); (Z.L.); (Z.Z.)
| | - Li Zhao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; (Y.L.); (X.D.); (L.S.); (H.C.); (J.K.); (N.B.); (Y.Z.); (Z.Q.); (Z.L.); (Z.Z.)
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture and Rural Affairs, Hohhot 010010, China
| |
Collapse
|
4
|
Nzelu CO, Bahrami S, Lawyer PG, Peters NC. Detection of Leishmania metacyclogenesis within the sand fly vector employing a real-time PCR for sherp gene expression: A tool for Leishmania surveillance and transmission potential. PLoS Negl Trop Dis 2025; 19:e0012915. [PMID: 40096086 PMCID: PMC11942415 DOI: 10.1371/journal.pntd.0012915] [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: 06/28/2024] [Revised: 03/26/2025] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Surveillance of infected insect vectors of vector-transmitted diseases has been recognized for its ability to estimate pathogen prevalence and transmission potential. Classically restricted to microscopic dissection and examination of individual insects, the potential of entomological monitoring has grown due to the advent of rapid molecular DNA detection methods with high specificity and sensitivity. Despite such advancement, a recurring question concerning DNA detection of parasitic pathogens is related to the fact that DNA amplification, by itself, does not differentiate between insects carrying infectious versus dead, non- or poorly-infectious life-cycle stages, thereby limiting it's programmatic usefulness for accurately measuring the transmission potential of infected insects in endemic areas or within experimentally infected populations. Herein, we developed a quantitative real-time PCR with Reverse Transcription (RT-qPCR) based sherp (small hydrophilic endoplasmic reticulum-associated protein) detection assay employing a novel set of sherp-RT-qPCR primers to detect and quantify infectious Leishmania parasites in infected vector sand flies. The sherp RT-qPCR showed significantly increased expression of sherp transcripts in infectious Leishmania metacyclic versus non-metacyclic promastigotes or mammalian-derived amastigotes. The assay displayed detection performance ranging from 106 to 1 parasite and could reliably quantify parasites within infected sand flies without the need for dissection. Sherp transcripts were also successfully amplified from flies stored in ethanol at room temperature, a practical and economical method of sample preservation in resource-limited field settings. Lastly, in conjunction with an established RT-qPCR assay for Leishmania kinetoplast DNA minicircles, we were able to calculate a score for the degree of metacyclogenesis within infected sand flies, a known predictor of transmission potential. These results highlight the potential of the sherp-RT-qPCR assay to identify hotspots of potential transmission, areas of re-emergence, vector competence, and the transmission potential of infected sand fly populations.
Collapse
Affiliation(s)
- Chukwunonso O. Nzelu
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, and Faculty of Veterinary Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Somayeh Bahrami
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, and Faculty of Veterinary Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Department of Parasitology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Phillip G. Lawyer
- Monte L. Bean Life Science Museum, Brigham Young University, MLBM, Provo, Utah, United States of America
| | - Nathan C. Peters
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, and Faculty of Veterinary Medicine, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
5
|
Vaselek S, Alten B. Microbial ecology of sandflies-the correlation between nutrition, Phlebotomus papatasi sandfly development and microbiome. Front Vet Sci 2025; 11:1522917. [PMID: 39911488 PMCID: PMC11794182 DOI: 10.3389/fvets.2024.1522917] [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: 11/06/2024] [Accepted: 12/16/2024] [Indexed: 02/07/2025] Open
Abstract
The role and the impact of the microbial component on the biology, ecology, and development of sandflies is largely unknown. We evaluated the impact of larval nutrition on laboratory-reared sandflies in correlation to the abundance of food, light starvation, and food with/without live microbiome, by monitoring the survival and development of immature stages, and the longevity of adult sandflies. Within this study we examined 360 larvae, 116 pupae, and 120 adult flies of Phlebotomus papatasi for the microbial gut content. The data showed that the presence of a live and diverse microbiome plays a role in the development and survival of larvae. The mortality rate of the larvae was higher, and larval development was longer for sandflies maintained on microbiome-depleted medium, in comparison to the larvae fed with medium containing alive and complex microbiome. Actively feeding larvae reduce microbial abundance and diversity of the medium. The microbial content of the larval gut depends on the composition of the rearing medium, indicating a potential attraction to certain bacteria. The microbial content of the pupa gut was severely diminished, with overall survival of two bacterial species in adult insects - Ochrobactrum intermedium (found in 95% of dissected adults) and Bacillus subtilis (16%). Further microbial studies may aid in developing biological control methods for sandfly larval or adult stages.
Collapse
Affiliation(s)
- Slavica Vaselek
- Department of Biology, Faculty of Science, Hacettepe University, Ankara, Türkiye
| | | |
Collapse
|
6
|
Duque-Granda D, Vivero-Gómez RJ, Junca H, Cadavid-Restrepo G, Moreno-Herrera CX. Interaction and effects of temperature preference under a controlled environment on the diversity and abundance of the microbiome in Lutzomyia longipalpis (Diptera: Psychodidae). BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2024; 44:e00857. [PMID: 39328926 PMCID: PMC11424975 DOI: 10.1016/j.btre.2024.e00857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 09/28/2024]
Abstract
Characterization of the temperature effects on the abundance and richness of the microbiota of Lutzomyia longipalpis, insect vector of Leishmania infantum in America, is an aspect of pivotal importance to understand the interactions between temperature, bacteria, and Leishmania infection. We developed and used a customized device with a temperature gradient (21-34 °C) to assess the temperature preferences of wild females of Lu. longipalpis collected in a rural area (Ricaurte, Cundinamarca, Colombia). Each replicate consisted of 50 females exposed to the gradient for an hour. At the end of the exposure time, insects were collected and separated by the temperature ranges selected varying from 21 °C to 34 °C. They were organized in 17 pools from which total DNA extracts were obtained, and samples were subjected to 16S rRNA amplicon sequencing analyzes. The most abundant phyla across the different temperature ranges were Proteobacteria (17.22-90.73 %), Firmicutes (5.99-77.21 %) and Actinobacteria (1.56-59.85 %). Results also showed an abundance (30 % to 57.36 %) of Pseudomonas (mainly at temperatures of 21-29 °C and 34 °C) that decreased to 6.55 %-13.20 % at temperatures of 31-33 °C, while Bacillus increase its abundance to 67.24 % at 29-33 °C. Serratia also had a greater representation (49.79 %), specifically in sand flies recovered at 25-27 °C. No significant differences were found at α-diversity level when comparing richness using the Shannon-Wiener, Simpson, and Chao1 indices, while β-diversity differences were found using the Bray-Curtis index (F-value of 3.5073, p-value < 0.013, R-squared of 0,4889), especially in the groups of Lu. longipalpis associated at higher temperatures (29-33 °C). It was also possible to detect the presence of endosymbionts such as Spiroplasma and Arsenophonus in the range of 29-33 °C. Rickettsia was only detected in Lu. longipalpis sand flies recovered between 25-27 °C. It was possible to characterize Lu. longipalpis microbiota in response to intraspecific temperature preferences and observe changes in bacterial communities and endosymbionts at different ranges of said environmental variable, which may be important in its vector competence and environmental plasticity to adapt to new climate change scenarios.
Collapse
Affiliation(s)
- Daniela Duque-Granda
- Grupo de Microbiodiversidad y Bioprospección, Laboratorio de Procesos Moleculares, Laboratorio de Biología Celular y Molecular, Universidad Nacional de Colombia sede Medellín, Street 59A #63-20, Medellín 050003, Colombia
| | - Rafael José Vivero-Gómez
- Grupo de Microbiodiversidad y Bioprospección, Laboratorio de Procesos Moleculares, Laboratorio de Biología Celular y Molecular, Universidad Nacional de Colombia sede Medellín, Street 59A #63-20, Medellín 050003, Colombia
| | - Howard Junca
- RG Microbial Ecology: Metabolism, Genomics & Evolution, Div. Ecogenomics & Holobionts, Microbiomas Foundation, LT11A, 250008, Chia, Colombia
| | - Gloria Cadavid-Restrepo
- Grupo de Microbiodiversidad y Bioprospección, Laboratorio de Procesos Moleculares, Laboratorio de Biología Celular y Molecular, Universidad Nacional de Colombia sede Medellín, Street 59A #63-20, Medellín 050003, Colombia
| | - Claudia Ximena Moreno-Herrera
- Grupo de Microbiodiversidad y Bioprospección, Laboratorio de Procesos Moleculares, Laboratorio de Biología Celular y Molecular, Universidad Nacional de Colombia sede Medellín, Street 59A #63-20, Medellín 050003, Colombia
| |
Collapse
|
7
|
Rogers ME, de Pablos LM, Sunter JD. Gels and cells: the Leishmania biofilm as a space and place for parasite transmission. Trends Parasitol 2024; 40:876-885. [PMID: 39218719 DOI: 10.1016/j.pt.2024.08.001] [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: 06/25/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Leishmania make an abundant glycoprotein and proteophosphoglycan-rich gel, called the promastigote secretory gel, in the anterior midgut of their sand fly vector. This gel is a multi-faceted virulence factor which promotes the survival and transmission of the parasites between hosts. Here, we present the case that Leishmania parasites embedded in the promastigote secretory gel should be redefined as a biofilm as it shares striking similarities in biogenesis, form, and function with biofilms of other unicellular organisms. We believe that this reinterpretation will stimulate new hypotheses and avenues of research to improve our understanding of the developmental programme of Leishmania and the interaction these parasites and other kinetoplastids have with their insect hosts.
Collapse
Affiliation(s)
- Matthew E Rogers
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, UK
| | - Luis Miguel de Pablos
- Department of Parasitology, University of Granada, Granada, Spain; Institute of Biotechnology, University of Granada, Granada, Spain
| | | |
Collapse
|
8
|
Tabbabi A, Mizushima D, Yamamoto DS, Zhioua E, Kato H. Comparative analysis of the microbiota of sand fly vectors of Leishmania major and L. tropica in a mixed focus of cutaneous leishmaniasis in southeast Tunisia; ecotype shapes the bacterial community structure. PLoS Negl Trop Dis 2024; 18:e0012458. [PMID: 39236074 PMCID: PMC11407667 DOI: 10.1371/journal.pntd.0012458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/17/2024] [Accepted: 08/14/2024] [Indexed: 09/07/2024] Open
Abstract
Phlebotomine sand flies are vectors of the protozoan parasite Leishmania spp. Although the intestinal microbiota is involved in a wide range of biological and physiological processes and has the potential to alter vector competence, little is known about the impact of host species and environment on the gut microbiome. To address this issue, a comparative analysis of the microbiota of sand fly vector populations of Leishmania major and L. tropica in a mixed focus of cutaneous leishmaniasis in Tunisia was performed. Bacterial 16S rRNA gene amplification and Illumina MiSeq sequencing were used to characterize and compare the overall bacterial and fungal composition of field-collected sand flies: Phlebotomus papatasi, Ph. perniciosus, Ph. riouxi, and Ph. sergenti. Thirty-eight bacterial genera belonging to five phyla were identified in 117 female specimens. The similarities and differences between the microbiome data from different samples collected from three collections were determined using principal coordinate analysis (PCoA). Substantial variations in the bacterial composition were found between geographically distinct populations of the same sand fly species, but not between different species at the same location, suggesting that the microbiota content was structured according to environmental factors rather than host species. These findings suggest that host phylogeny may play a minor role in determining the insect gut microbiota, and its potential to affect the transmission of the Leishmania parasite appear to be very low. These results highlight the need for further studies to decode sand fly Leishmania-microbiota interactions, as even the same bacterial species, such as Enterococcus faecalis, can exert completely opposite effects when confronted with different pathogens within various host insects and vice versa.
Collapse
Affiliation(s)
- Ahmed Tabbabi
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Daiki Mizushima
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Daisuke S Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| | - Elyes Zhioua
- Unit of Vector Ecology, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Hirotomo Kato
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
9
|
Vomáčková Kykalová B, Sassù F, Dutra-Rêgo F, Soares RP, Volf P, Loza Telleria E. Pathogen-associated molecular patterns (PAMPs) derived from Leishmania and bacteria increase gene expression of antimicrobial peptides and gut surface proteins in sand flies. Int J Parasitol 2024; 54:485-495. [PMID: 38626865 DOI: 10.1016/j.ijpara.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/12/2024] [Accepted: 04/10/2024] [Indexed: 04/30/2024]
Abstract
The interaction between pathogens and vectors' physiology can impact parasite transmission. Studying this interaction at the molecular level can help in developing control strategies. We study leishmaniases, diseases caused by Leishmania parasites transmitted by sand fly vectors, posing a significant global public health concern. Lipophosphoglycan (LPG), the major surface glycoconjugate of Leishmania, has been described to have several roles throughout the parasite's life cycle, both in the insect and vertebrate hosts. In addition, the sand fly midgut possesses a rich microbiota expressing lipopolysaccharides (LPS). However, the effect of LPG and LPS on the gene expression of sand fly midgut proteins or immunity effectors has not yet been documented. We experimentally fed Lutzomyia longipalpis and Phlebotomus papatasi sand flies with blood containing purified LPG from Leishmania infantum, Leishmania major, or LPS from Escherichia coli. The effect on the expression of genes encoding gut proteins galectin and mucin, digestive enzymes trypsin and chymotrypsin, and antimicrobial peptides (AMPs) attacin and defensins was assessed by quantitative PCR (qPCR). The gene expression of a mucin-like protein in L. longipalpis was increased by L. infantum LPG and E. coli LPS. The gene expression of a galectin was increased in L. longipalpis by L. major LPG, and in P. papatasi by E. coli LPS. Nevertheless, the gene expression of trypsins and chymotrypsins did not significantly change. On the other hand, both L. infantum and L. major LPG significantly enhanced expression of the AMP attacin in both sand fly species and defensin in L. longipalpis. In addition, E. coli LPS increased the expression of attacin and defensin in L. longipalpis. Our study showed that Leishmania LPG and E. coli LPS differentially modulate the expression of sand fly genes involved in gut maintenance and defence. This suggests that the glycoconjugates from microbiota or Leishmania may increase the vector's immune response and the gene expression of a gut coating protein in a permissive vector.
Collapse
Affiliation(s)
- Barbora Vomáčková Kykalová
- Charles University, Faculty of Science, Department of Parasitology, Viničná 7, 128 44, Prague, Czech Republic
| | - Fabiana Sassù
- Charles University, Faculty of Science, Department of Parasitology, Viničná 7, 128 44, Prague, Czech Republic
| | - Felipe Dutra-Rêgo
- Biotechnology Applied to Pathogens (BAP), Instituto René Rachou, Fundação Oswaldo Cruz (Fiocruz), Av. Augusto de Lima, 1715, CEP: 30190-009, Belo Horizonte, MG, Brazil
| | - Rodrigo Pedro Soares
- Biotechnology Applied to Pathogens (BAP), Instituto René Rachou, Fundação Oswaldo Cruz (Fiocruz), Av. Augusto de Lima, 1715, CEP: 30190-009, Belo Horizonte, MG, Brazil
| | - Petr Volf
- Charles University, Faculty of Science, Department of Parasitology, Viničná 7, 128 44, Prague, Czech Republic
| | - Erich Loza Telleria
- Charles University, Faculty of Science, Department of Parasitology, Viničná 7, 128 44, Prague, Czech Republic.
| |
Collapse
|
10
|
Layouni S, Remadi L, Kidar A, Chaâbane-Banaoues R, Haouas N, Babba H. Clinical polymorphism of zoonotic cutaneous leishmaniasis: combination of the clinical and the parasitological diagnosis. Parasitol Res 2024; 123:238. [PMID: 38856772 DOI: 10.1007/s00436-024-08263-5] [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: 02/09/2024] [Accepted: 06/02/2024] [Indexed: 06/11/2024]
Abstract
Zoonotic cutaneous leishmaniasis (ZCL) is a neglected tropical disease caused by Leishmania (L.) major. This zoonosis is characterized by a broad-spectrum clinical polymorphism and may be underestimated and poorly treated since it is a simulator of various dermatoses. The aim of our study was to analyze the clinical polymorphism of patients with ZCL. A total of 142 patients with confirmed CL based on the microscopic examination of skin lesion biopsies were included in this study. Molecular typing of Leishmania species revealed that all patients were infected with L. major. In total, 14 clinical forms were observed. Six were typical and eight were atypical. The typical ZCL forms are grouped as follows: papular (26.76%), ulcero-crusted (26.05%), ulcerated (13.38%), impetiginous (9.86%), nodular (9.15%), and papulo-nodular (5.63%) lesions. In atypical ZCL forms, we described erythematous (2.81%), erysipeloid (1.4%), sporotrichoid, (1.4%), keratotic (0.7%) lupoid (0.7%), lichenoid (0.7%), psoriasiform (0.7%), and zosteriform (0.7%) lesions. Here, the lichenoid and the keratotic forms caused by L. major were reported for the first time in Tunisia. These findings will help physicians to be aware of the unusual lesions of ZCL that could be confused with other dermatological diseases. For this reason, it will be necessary to improve the diagnosis of CL especially in endemic areas. Such large clinical polymorphism caused by L. major may be the result of a complex association between the vector microbiota, the parasite, and the host immune state, and further studies should be carried out in order to reveal the mechanisms involved in clinical polymorphism of ZCL.
Collapse
Affiliation(s)
- Samia Layouni
- Laboratory of Medical and Molecular Parasitology-Mycology LP3M (Code LR12ES08), Department of Clinical Biology B, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia.
- Department of Nutrition and Environmental Sciences, Higher Institute of Applied Sciences and Technology of Mahdia, University of Monastir, Monastir, Tunisia.
| | - Latifa Remadi
- Laboratory of Medical and Molecular Parasitology-Mycology LP3M (Code LR12ES08), Department of Clinical Biology B, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
- Laboratory of Molecular Entomology, Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, Heraklion, Greece
| | | | - Raja Chaâbane-Banaoues
- Laboratory of Medical and Molecular Parasitology-Mycology LP3M (Code LR12ES08), Department of Clinical Biology B, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Najoua Haouas
- Laboratory of Medical and Molecular Parasitology-Mycology LP3M (Code LR12ES08), Department of Clinical Biology B, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Hamouda Babba
- Laboratory of Medical and Molecular Parasitology-Mycology LP3M (Code LR12ES08), Department of Clinical Biology B, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| |
Collapse
|
11
|
Vivero-Gomez R, Duque-Granda D, Rader JA, Stuckert A, Santander-Gualdron R, Cadavid-Restrepo G, Moreno-Herrera CX, Matute DR. Humidity and temperature preference in two Neotropical species of sand flies. Parasit Vectors 2024; 17:246. [PMID: 38831449 PMCID: PMC11149334 DOI: 10.1186/s13071-024-06325-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 05/15/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Arthropods vector a multitude of human disease-causing organisms, and their geographic ranges are shifting rapidly in response to changing climatic conditions. This is, in turn, altering the landscape of disease risk for human populations that are brought into novel contact with the vectors and the diseases they carry. Sand flies in the genera Lutzomyia and Pintomyia are vectors of serious disease-causing agents such as Leishmania (the etiological agent of leishmaniasis) and may be expanding their range in the face of climate change. Understanding the climatic conditions that vector species both tolerate physiologically and prefer behaviorally is critical to predicting the direction and magnitude of range expansions and the resulting impacts on human health. Temperature and humidity are key factors that determine the geographic extent of many arthropods, including vector species. METHODS We characterized the habitat of two species of sand flies, Lutzomyia longipalpis and Pintomyia evansi. Additionally, we studied two behavioral factors of thermal fitness-thermal and humidity preference in two species of sand flies alongside a key aspect of physiological tolerance-desiccation resistance. RESULTS We found that Lu. longipalpis is found at cooler and drier conditions than Pi. evansi. Our results also show significant interspecific differences in both behavioral traits, with Pi. evansi preferring warmer, more humid conditions than Lu. longipalpis. Finally, we found that Lu. longipalpis shows greater tolerance to extreme low humidity, and that this is especially pronounced in males of the species. CONCLUSIONS Taken together, our results suggest that temperature and humidity conditions are key aspects of the climatic niche of Lutzomyia and Pintomyia sand flies and underscore the value of integrative studies of climatic tolerance and preference in vector biology.
Collapse
Affiliation(s)
- Rafael Vivero-Gomez
- Grupo de Microdiversidad and Bioprospección, Facultad de Ciencias, Departamento de Biociencias, Laboratorio de Procesos Moleculares, Universidad Nacional de Colombia, Sede Medellín, Medellín, Colombia
- PECET (Programa de Estudio y Control de Enfermedades Tropicales), Universidad de Antioquia, SIU-Sede de Investigación Universitaria, Street 62 # 52-59Laboratory 632, 050003, Medellín, Postal Code, Colombia
| | - Daniela Duque-Granda
- Grupo de Microdiversidad and Bioprospección, Facultad de Ciencias, Departamento de Biociencias, Laboratorio de Procesos Moleculares, Universidad Nacional de Colombia, Sede Medellín, Medellín, Colombia
| | - Jonathan A Rader
- Biology Department, University of North Carolina, Chapel Hill, USA
| | - Adam Stuckert
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Ricardo Santander-Gualdron
- Grupo de Microdiversidad and Bioprospección, Facultad de Ciencias, Departamento de Biociencias, Laboratorio de Procesos Moleculares, Universidad Nacional de Colombia, Sede Medellín, Medellín, Colombia
| | - Gloria Cadavid-Restrepo
- Grupo de Microdiversidad and Bioprospección, Facultad de Ciencias, Departamento de Biociencias, Laboratorio de Procesos Moleculares, Universidad Nacional de Colombia, Sede Medellín, Medellín, Colombia
| | - Claudia X Moreno-Herrera
- Grupo de Microdiversidad and Bioprospección, Facultad de Ciencias, Departamento de Biociencias, Laboratorio de Procesos Moleculares, Universidad Nacional de Colombia, Sede Medellín, Medellín, Colombia
| | - Daniel R Matute
- Biology Department, University of North Carolina, Chapel Hill, USA.
| |
Collapse
|
12
|
Guo W, Zhu W, Jia L, Tao Y. Unique microbial communities of parasitic fleas on wild animals from the Qinghai-Tibet Plateau. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:40916-40924. [PMID: 38834927 DOI: 10.1007/s11356-024-33885-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 05/29/2024] [Indexed: 06/06/2024]
Abstract
Fleas, one of the most significant ectoparasites, play a crucial role as vectors in spreading zoonotic diseases globally. The Qinghai Province, as part of the Qinghai-Tibet Plateau, is one of the provinces in China with the largest number of flea species. In this study, we characterized the microbial communities of eighty-five adult fleas, belonging to nineteen species within four families (Ceratophyllidae, Ctenophthalmidae, Leptopsyllidae, and Pulicidae). We identified a total of 1162 unique operational taxonomic units at the genus level, with flea-borne pathogens such as Wolbachia, Bartonella, Rickettsia being the members of top abundant taxa. Except for comparison between Ctenophthalmidae and Leptopsyllidae families, the analyses of both alpha- and beta- diversity indicators suggested that bacterial diversity varied among flea families. This could be attributed to flea phylogeny, which also influenced by their geographical sites and animal hosts. Results of Linear discriminant analysis effect size (LEfSe) indicated that 29 genera in Ceratophylloidea, 11 genera in Ctenophthalmidae, 15 genera in Leptopsyllidae, and 22 genera in Pulicidae were significantly responsible for explaining the differences among the four flea families (linear discriminant analysis score > 2, P < 0.05). Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt2) analyses showed that the functional pathways varied significantly across flea families, which was supported by the significant correlation between the functional pathways and the microbial communities.
Collapse
Affiliation(s)
- Wentao Guo
- Qinghai Institute for Endemic Disease Prevention and Control, Xining, 811602, China
| | - Wentao Zhu
- Department of Infectious Diseases and Clinical Microbiology, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Luo Jia
- Qinghai Institute for Endemic Disease Prevention and Control, Xining, 811602, China
| | - Yuanqing Tao
- Qinghai Institute for Endemic Disease Prevention and Control, Xining, 811602, China.
| |
Collapse
|
13
|
Ratcliffe NA, Mello CB, Castro HC, Dyson P, Figueiredo M. Immune Reactions of Vector Insects to Parasites and Pathogens. Microorganisms 2024; 12:568. [PMID: 38543619 PMCID: PMC10974449 DOI: 10.3390/microorganisms12030568] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 11/12/2024] Open
Abstract
This overview initially describes insect immune reactions and then brings together present knowledge of the interactions of vector insects with their invading parasites and pathogens. It is a way of introducing this Special Issue with subsequent papers presenting the latest details of these interactions in each particular group of vectors. Hopefully, this paper will fill a void in the literature since brief descriptions of vector immunity have now been brought together in one publication and could form a starting point for those interested and new to this important area. Descriptions are given on the immune reactions of mosquitoes, blackflies, sandflies, tsetse flies, lice, fleas and triatomine bugs. Cellular and humoral defences are described separately but emphasis is made on the co-operation of these processes in the completed immune response. The paper also emphasises the need for great care in extracting haemocytes for subsequent study as appreciation of their fragile nature is often overlooked with the non-sterile media, smearing techniques and excessive centrifugation sometimes used. The potential vital role of eicosanoids in the instigation of many of the immune reactions described is also discussed. Finally, the priming of the immune system, mainly in mosquitoes, is considered and one possible mechanism is presented.
Collapse
Affiliation(s)
- Norman Arthur Ratcliffe
- Department of Biosciences, Swansea University, Singleton Park, Swansea SA28PP, UK
- Biology Institute, Universidade Federal Fluminense, Niterói 24210-130, RJ, Brazil; (C.B.M.); (H.C.C.)
| | - Cicero Brasileiro Mello
- Biology Institute, Universidade Federal Fluminense, Niterói 24210-130, RJ, Brazil; (C.B.M.); (H.C.C.)
| | - Helena Carla Castro
- Biology Institute, Universidade Federal Fluminense, Niterói 24210-130, RJ, Brazil; (C.B.M.); (H.C.C.)
| | - Paul Dyson
- Institute of Life Science, Medical School, Swansea University, Singleton Park, Swansea SA28PP, UK; (P.D.); (M.F.)
| | - Marcela Figueiredo
- Institute of Life Science, Medical School, Swansea University, Singleton Park, Swansea SA28PP, UK; (P.D.); (M.F.)
| |
Collapse
|
14
|
Mrázek J, Mrázková L, Mekadim C, Jarošíková T, Krayem I, Sohrabi Y, Demant P, Lipoldová M. Effects of Leishmania major infection on the gut microbiome of resistant and susceptible mice. Appl Microbiol Biotechnol 2024; 108:145. [PMID: 38240984 PMCID: PMC10799115 DOI: 10.1007/s00253-024-13002-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 12/22/2023] [Accepted: 01/03/2024] [Indexed: 01/22/2024]
Abstract
Cutaneous leishmaniasis, a parasitic disease caused by Leishmania major, is a widely frequent form in humans. To explore the importance of the host gut microbiota and to investigate its changes during L. major infection, two different groups of mouse models were assessed. The microbiome of two parts of the host gut-ileum and colon-from infected and non-infected mice were characterised by sequencing of 16S rDNA using an Ion Torrent PGM platform. Microbiome analysis was performed to reveal changes related to the susceptibility and the genetics of mice strains in two different gut compartments and to compare the results between infected and non-infected mice. The results showed that Leishmania infection affects mainly the ileum microbiota, whereas the colon bacterial community was more stable. Different biomarkers were determined in the gut microbiota of infected resistant mice and infected susceptible mice using LEfSe analysis. Lactobacillaceae was associated with resistance in the colon microbiota of all resistant mice strains infected with L. major. Genes related to xenobiotic biodegradation and metabolism and amino acid metabolism were primarily enriched in the small intestine microbiome of resistant strains, while genes associated with carbohydrate metabolism and glycan biosynthesis and metabolism were most abundant in the gut microbiome of the infected susceptible mice. These results should improve our understanding of host-parasite interaction and provide important insights into the effect of leishmaniasis on the gut microbiota. Also, this study highlights the role of host genetic variation in shaping the diversity and composition of the gut microbiome. KEY POINTS: • Leishmaniasis may affect mainly the ileum microbiota while colon microbiota was more stable. • Biomarkers related with resistance or susceptibility were determined in the gut microbiota of mice. • Several pathways were predicted to be upregulated in the gut microbiota of resistant or susceptible mice.
Collapse
Affiliation(s)
- Jakub Mrázek
- Laboratory of Anaerobic Microbiology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic
| | - Lucie Mrázková
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic
- Faculty of Biomedical Engineering, Czech Technical University in Prague, Namestí Sitna 3105, 272 01, Kladno, Czech Republic
| | - Chahrazed Mekadim
- Laboratory of Anaerobic Microbiology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic.
| | - Taťána Jarošíková
- Faculty of Biomedical Engineering, Czech Technical University in Prague, Namestí Sitna 3105, 272 01, Kladno, Czech Republic
| | - Imtissal Krayem
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic
| | - Yahya Sohrabi
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic
- Department of Medical Genetics, 3Rd Faculty of Medicine, Charles University, Ruská 87, 100 00, Prague 10, Czech Republic
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Westfälische Wilhelms-Universität, Münster, Germany
| | - Peter Demant
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Marie Lipoldová
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague, Czech Republic
- Faculty of Biomedical Engineering, Czech Technical University in Prague, Namestí Sitna 3105, 272 01, Kladno, Czech Republic
- Department of Medical Genetics, 3Rd Faculty of Medicine, Charles University, Ruská 87, 100 00, Prague 10, Czech Republic
| |
Collapse
|
15
|
Tom A, Kumar NP, Kumar A, Saini P. Interactions between Leishmania parasite and sandfly: a review. Parasitol Res 2023; 123:6. [PMID: 38052752 DOI: 10.1007/s00436-023-08043-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 11/06/2023] [Indexed: 12/07/2023]
Abstract
Leishmaniasis transmission cycles are maintained and sustained in nature by the complex crosstalk of the Leishmania parasite, sandfly vector, and the mammalian hosts (human, as well as zoonotic reservoirs). Regardless of the vast research on human host-parasite interaction, there persists a substantial knowledge gap on the parasite's development and modulation in the vector component. This review focuses on some of the intriguing aspects of the Leishmania-sandfly interface, beginning with the uptake of the intracellular amastigotes from an infected host to the development of the parasite within the sandfly's alimentary canal, followed by the transmission of infective metacyclic stages to another potential host. Upon ingestion of the parasite, the sandfly hosts an intricate repertoire of immune barriers, either to evade the parasite or to ensure its homeostatic coexistence with the vector gut microbiome. Sandfly salivary polypeptides and Leishmania exosomes are co-egested with the parasite inoculum during the infected vector bite. This has been attributed to the modulation of the parasite infection and subsequent clinical manifestation in the host. While human host-based studies strive to develop effective therapeutics, a greater understanding of the vector-parasite-microbiome and human host interactions could help us to identify the targets and to develop strategies for effectively preventing the transmission of leishmaniasis.
Collapse
Affiliation(s)
- Anns Tom
- ICMR-Vector Control Research Centre (Field Station), Kottayam, Kerala, India
| | - N Pradeep Kumar
- ICMR-Vector Control Research Centre (Field Station), Kottayam, Kerala, India
| | - Ashwani Kumar
- ICMR- Vector Control Research Centre, Puducherry, India
| | - Prasanta Saini
- ICMR-Vector Control Research Centre (Field Station), Kottayam, Kerala, India.
| |
Collapse
|
16
|
Swanson K, Blakeslee AMH, Fowler AE, Roozbehi S, Field EK. Microbial communities are indicators of parasite infection status. Environ Microbiol 2023; 25:3423-3434. [PMID: 37918974 DOI: 10.1111/1462-2920.16533] [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: 04/13/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023]
Abstract
Growing evidence suggests that microbiomes have been shaping the evolutionary pathways of macroorganisms for millennia and that these tiny symbionts can influence, and possibly even control, species interactions like host-parasite relationships. Yet, while studies have investigated host-parasites and microbiomes separately, little has been done to understand all three groups synergistically. Here, we collected infected and uninfected Eurypanopeus depressus crab hosts from a coastal North Carolina oyster reef three times over 4 months. Infected crabs demonstrated an external stage of the rhizocephalan parasite, Loxothylacus panopaei. Community analyses revealed that microbial richness and diversity were significantly different among tissue types (uninfected crab, infected crab, parasite externae and parasite larvae) and over time (summer and fall). Specifically, the microbial communities from parasite externae and larvae had similar microbiomes that were consistent through time. Infected crabs demonstrated microbial communities spanning those of their host and parasite, while uninfected crabs showed more distinctive communities with greater variability over time. Microbial communities were also found to be indicators of early-stage infections. Resolving the microbial community composition of a host and its parasite is an important step in understanding the microbiome's role in the host-parasite relationship and determining how this tripartite relationship impacts coevolutionary processes.
Collapse
Affiliation(s)
- Kyle Swanson
- Department of Biology, East Carolina University, Greenville, North Carolina, USA
| | - April M H Blakeslee
- Department of Biology, East Carolina University, Greenville, North Carolina, USA
| | - Amy E Fowler
- Environmental Science & Policy Department, George Mason University, Fairfax, Virginia, USA
| | - Sara Roozbehi
- Department of Biology, East Carolina University, Greenville, North Carolina, USA
| | - Erin K Field
- Department of Biology, East Carolina University, Greenville, North Carolina, USA
| |
Collapse
|
17
|
Abstract
Haematophagous arthropods, including mosquitoes, ticks, flies, triatomine bugs and lice (here referred to as vectors), are involved in the transmission of various pathogens to mammals on whom they blood feed. The diseases caused by these pathogens, collectively known as vector-borne diseases (VBDs), threaten the health of humans and animals. Although the vector arthropods differ in life histories, feeding behaviour as well as reproductive strategies, they all harbour symbiotic microorganisms, known as microbiota, on which they depend for completing essential aspects of their biology, such as development and reproduction. In this Review, we summarize the shared and unique key features of the symbiotic associations that have been characterized in the major vector taxa. We discuss the crosstalks between microbiota and their arthropod hosts that influence vector metabolism and immune responses relevant for pathogen transmission success, known as vector competence. Finally, we highlight how current knowledge on symbiotic associations is being explored to develop non-chemical-based alternative control methods that aim to reduce vector populations, or reduce vector competence. We conclude by highlighting the remaining knowledge gaps that stand to advance basic and translational aspects of vector-microbiota interactions.
Collapse
Affiliation(s)
- Jingwen Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, P. R. China.
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, P. R. China.
| | - Li Gao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, P. R. China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, P. R. China
| | - Serap Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, CT, USA
| |
Collapse
|
18
|
Naz S, Ali Z, Minhas A, Fatima A, Waseem S. Generation of dysbiotic microbiota in cutaneous Leishmaniasis and enhancement of skin inflammation. Microb Pathog 2023; 181:106202. [PMID: 37327948 DOI: 10.1016/j.micpath.2023.106202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/18/2023]
Abstract
Cutaneous Leishmaniasis (CL) affects millions of people globally and has a significant impact on morbidity and mortality. Innate immune mediators are likely to influence the clinical phenotype of CL through primary responses that restrict or facilitate parasite spread. The aim of the study was to bring to attention the significance of microbiota in the development of CL and emphasized the necessity of including the role of microbiota in CL while promoting a One Health approach for managing diseases. To achieve this, we used 16S amplicon metagenome sequencing and QIIME2 pipeline to analyze the microbiome composition of CL-infected patients compared to non-infected, healthy subjects. 16S sequencing analysis showed serum microbiome was dominated by Firmicutes, Proteobacteria, Bacteroidota, and Actinobacteria. CL-infected individuals, Proteobacteria were the most prevalent (27.63 ± 9.79), with the relative abundance (10.73 ± 5.33) of Proteobacteria in control. Bacilli class was found to be the most prevalent in healthy controls (30.71 ± 8.44) while (20.57 ± 9.51) in CL-infected individuals. The class Alphaproteobacteria was found to be more in CL-infected individuals (5.47 ± 2.07) as compared to healthy controls (1.85 ± 0.39). The CL-infected individuals had a significantly lower relative abundance of the Clostridia class (p < 0.0001). An altered serum microbiome of CL infection and higher microbial abundance in the serum of healthy individuals was observed.
Collapse
Affiliation(s)
- Shumaila Naz
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Rawalpindi, 46000, Pakistan.
| | - Zain Ali
- ABO SCIENTIFIC, Chakri Road, Rawalpindi, Pakistan
| | - Azhar Minhas
- Department of Dermatology, Combined Military Hospital (CMH), Quetta, 87300, Pakistan
| | - Anam Fatima
- Department of Medicine, Polyclinic Hospital, Islamabad, Pakistan
| | | |
Collapse
|
19
|
Vomáčková Kykalová B, Sassù F, Volf P, Telleria EL. RNAi-mediated gene silencing of Phlebotomus papatasi defensins favors Leishmania major infection. Front Physiol 2023; 14:1182141. [PMID: 37265840 PMCID: PMC10230645 DOI: 10.3389/fphys.2023.1182141] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/25/2023] [Indexed: 06/03/2023] Open
Abstract
Introduction: Production of different antimicrobial peptides (AMPs) is one of the insect's prominent defense strategies, regulated mainly by Toll and immune deficiency (IMD) humoral pathways. Here we focused mainly on two AMPs of Phlebotomus papatasi, vector of Leishmania major parasites, their association with the relish transcription factor and the effective participation on Leishmania infection. Methods and results: We further characterized the role of previously described gut-specific P. papatasi defensin (PpDef1) and identified the second defensin (PpDef2) expressed in various sand fly tissues. Using the RNAi-mediated gene silencing, we report that the silencing of PpDef1 gene or simultaneous silencing of both defensin genes (PpDef1 and PpDef2) resulted in increased parasite levels in the sand fly (detectable by PCR) and higher sand fly mortality. In addition, we knocked down relish, the sole transcription factor of the IMD pathway, to evaluate the association of the IMD pathway with AMPs expression in P. papatasi. We demonstrated that the relish gene knockdown reduced the expression of PpDef2 and attacin, another AMP abundantly expressed in the sand fly body. Conclusions: Altogether, our experiments show the importance of defensins in the sand fly response toward L. major and the role of the IMD pathway in regulating AMPs in P. papatasi.
Collapse
|
20
|
Lozano-Sardaneta YN, Marina CF, Torres-Monzón JA, Sánchez-Cordero V, Becker I. Molecular detection of Wolbachia and Bartonella as part of the microbiome of phlebotomine sand flies from Chiapas, Mexico. Parasitol Res 2023; 122:1293-1301. [PMID: 37055642 PMCID: PMC10172221 DOI: 10.1007/s00436-023-07829-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/24/2023] [Indexed: 04/15/2023]
Abstract
Phlebotomine sand flies are dipterans of relevance due to their role as vectors of several pathogens worldwide. Bacteria in the gut of sand flies possibly affect their vectorial capacity and competence to transmit parasites. A retrospective study was performed in sand fly specimens that had previously been collected in four localities of the state of Chiapas during the period 2009-2011 to detect Wolbachia and Bartonella and their possible coinfection with Leishmania. For the molecular detection of bacteria, we used primers and conditions that had previously been reported. A total of 531 sand fly specimens of 10 species were analyzed. Four Wolbachia strains were detected in five sand fly species, showing a prevalence of 8.6%. All the Wolbachia strains had previously been reported in other taxa. In one sand fly species, we also detected a new lineage of Bartonella evidenced by a phylogenetic analysis. No sand fly specimens showed coinfections of these bacteria and Leishmania. The bacteria found in the phlebotomine sand flies are possibly transmitted by plant-mediated horizontal transmission and during blood meal feeding.
Collapse
Affiliation(s)
- Yokomi N Lozano-Sardaneta
- Centro de Medicina Tropical, Unidad de Medicina Experimental, Facultad de Medicina de la Universidad Nacional Autónoma de México, Hospital General de México, Dr. Balmis 148, Col. Doctores, 06726, Mexico City, México
| | - Carlos F Marina
- Centro Regional de Investigación en Salud Pública, Instituto Nacional de Salud Pública (CRISP-INSP), Tapachula, Chiapas, México
| | - Jorge A Torres-Monzón
- Centro Regional de Investigación en Salud Pública, Instituto Nacional de Salud Pública (CRISP-INSP), Tapachula, Chiapas, México
| | - Víctor Sánchez-Cordero
- Instituto de Biología, Universidad Nacional Autónoma de México, Ciudad de Mexico, 04510, México
| | - Ingeborg Becker
- Centro de Medicina Tropical, Unidad de Medicina Experimental, Facultad de Medicina de la Universidad Nacional Autónoma de México, Hospital General de México, Dr. Balmis 148, Col. Doctores, 06726, Mexico City, México.
| |
Collapse
|
21
|
Layouni S, Remadi L, Chaâbane-Banaoues R, Haouas N, Babba H. Identification of cuticle and midgut fungal microflora of phlebotomine sandflies collected in Tunisia. Arch Microbiol 2023; 205:64. [PMID: 36633698 DOI: 10.1007/s00203-022-03386-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023]
Abstract
Phlebotomine sand flies (Diptera: Psychodidae) are the proven vectors of Leishmaniases which are widespread parasitosis in many tropical and subtropical countries. The development of infective metacyclic Leishmania (Kinetoplastida: Trypanosomatidae) promastigotes stage is restricted to the vector midgut. Recently, several studies have assessed the influence of the sand fly midgut fungal microflora on the development of invective Leishmania stage. The aim of this study was to identify the fungal microflora from the cuticle and midgut of wild caught sandflies. A total of 50 sandflies were caught in two different leishmaniasis foci of center Tunisia and analyzed using an in vitro isolation of fungi followed by a morphological and molecular identification of fungal isolates. The morphological identification of sandflies specimens revealed five Species: Phlebotomus (P.) papatasi (n = 25), P. perniciosus (n = 15) P. riouxi (n = 6), P. longicuspis (n = 3) and P. sergenti (n = 1). Forty positive fungal cultures were isolated from 34 sand flies (19 males and 15 females) distributed as following: P. papatasi (n = 16), P. perniciosus (n = 11), P. riouxi (n = 4), P. longicuspis (n = 2) and P. sergenti (n = 1). Thirty-five cultures were isolated from the cuticles and five from the guts. A total of 15 fungi genera belonging to 8 families were identified with the predominance of Aspergillus genus followed by Penicillium genus. Among the 15 fungi genera, five were common between males and females specimens. Lecytophora canina and Leishmania major co-infection was detected in the gut of a female P. papatasi. Our preliminary findings highlight the high diversity of fungal microflora from the sand flies midguts.
Collapse
Affiliation(s)
- Samia Layouni
- Laboratory of Medical and Molecular Parasitology-Mycology LP3M (Code LR12ES08), Department of Clinical Biology B, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia. .,Department of Nutrition and Environmental Sciences, Higher Institute of Applied Sciences and Technology of Mahdia, University of Monastir, Monastir, Tunisia.
| | - Latifa Remadi
- Laboratory of Medical and Molecular Parasitology-Mycology LP3M (Code LR12ES08), Department of Clinical Biology B, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Raja Chaâbane-Banaoues
- Laboratory of Medical and Molecular Parasitology-Mycology LP3M (Code LR12ES08), Department of Clinical Biology B, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Najoua Haouas
- Laboratory of Medical and Molecular Parasitology-Mycology LP3M (Code LR12ES08), Department of Clinical Biology B, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Hamouda Babba
- Laboratory of Medical and Molecular Parasitology-Mycology LP3M (Code LR12ES08), Department of Clinical Biology B, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| |
Collapse
|
22
|
Amni F, Maleki-Ravasan N, Nateghi-Rostami M, Hadighi R, Karimian F, Meamar AR, Badirzadeh A, Parvizi P. Co-infection of Phlebotomus papatasi (Diptera: Psychodidae) gut bacteria with Leishmania major exacerbates the pathological responses of BALB/c mice. Front Cell Infect Microbiol 2023; 13:1115542. [PMID: 36779192 PMCID: PMC9909354 DOI: 10.3389/fcimb.2023.1115542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
Clinical features and severity of the leishmaniasis is extremely intricate and depend on several factors, especially sand fly-derived products. Bacteria in the sand fly's gut are a perpetual companion of Leishmania parasites. However, consequences of the concomitance of these bacteria and Leishmania parasite outside the midgut environment have not been investigated in the infection process. Herein, a needle infection model was designed to mimic transmission by sand flies, to examine differences in the onset and progression of L. major infection initiated by inoculation with "low" or "high" doses of Enterobacter cloacae and Bacillus subtilis bacteria. The results showed an alteration in the local expression of pro- and anti-inflammatory cytokines in mice receiving different inoculations of bacteria. Simultaneous injection of two bacteria with Leishmania parasites in the low-dose group caused greater thickness of ear pinna and enhanced tissue chronic inflammatory cells, as well as resulted in multifold increase in the expression of IL-4 and IL-1β and a decrease in the iNOS expression, without changing the L. major burden. Despite advances in scientific breakthroughs, scant survey has investigated the interaction between micro and macro levels of organization of leishmaniasis that ranges from the cellular to macro ecosystem levels, giving rise to the spread and persistence of the disease in a region. Our findings provide new insight into using the potential of the vector-derived microbiota in modulating the vertebrate immune system for the benefit of the host or recommend the use of appropriate antibiotics along with antileishmanial medicines.
Collapse
Affiliation(s)
- Fariba Amni
- Department of Parasitology and Mycology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Naseh Maleki-Ravasan
- Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
- *Correspondence: Naseh Maleki-Ravasan, ; Mahmoud Nateghi-Rostami, ; Ramtin Hadighi, ; Parviz Parvizi,
| | - Mahmoud Nateghi-Rostami
- Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
- *Correspondence: Naseh Maleki-Ravasan, ; Mahmoud Nateghi-Rostami, ; Ramtin Hadighi, ; Parviz Parvizi,
| | - Ramtin Hadighi
- Department of Parasitology and Mycology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- *Correspondence: Naseh Maleki-Ravasan, ; Mahmoud Nateghi-Rostami, ; Ramtin Hadighi, ; Parviz Parvizi,
| | - Fateh Karimian
- Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
| | - Ahmad Reza Meamar
- Department of Parasitology and Mycology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Badirzadeh
- Department of Parasitology and Mycology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parviz Parvizi
- Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
- *Correspondence: Naseh Maleki-Ravasan, ; Mahmoud Nateghi-Rostami, ; Ramtin Hadighi, ; Parviz Parvizi,
| |
Collapse
|
23
|
Trzebny A, Slodkowicz-Kowalska A, Björkroth J, Dabert M. Microsporidian Infection in Mosquitoes (Culicidae) Is Associated with Gut Microbiome Composition and Predicted Gut Microbiome Functional Content. MICROBIAL ECOLOGY 2023; 85:247-263. [PMID: 34939130 PMCID: PMC9849180 DOI: 10.1007/s00248-021-01944-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 12/10/2021] [Indexed: 06/14/2023]
Abstract
The animal gut microbiota consist of many different microorganisms, mainly bacteria, but archaea, fungi, protozoans, and viruses may also be present. This complex and dynamic community of microorganisms may change during parasitic infection. In the present study, we investigated the effect of the presence of microsporidians on the composition of the mosquito gut microbiota and linked some microbiome taxa and functionalities to infections caused by these parasites. We characterised bacterial communities of 188 mosquito females, of which 108 were positive for microsporidian DNA. To assess how bacterial communities change during microsporidian infection, microbiome structures were identified using 16S rRNA microbial profiling. In total, we identified 46 families and four higher taxa, of which Comamonadaceae, Enterobacteriaceae, Flavobacteriaceae and Pseudomonadaceae were the most abundant mosquito-associated bacterial families. Our data suggest that the mosquito gut microbial composition varies among host species. In addition, we found a correlation between the microbiome composition and the presence of microsporidians. The prediction of metagenome functional content from the 16S rRNA gene sequencing suggests that microsporidian infection is characterised by some bacterial species capable of specific metabolic functions, especially the biosynthesis of ansamycins and vancomycin antibiotics and the pentose phosphate pathway. Moreover, we detected a positive correlation between the presence of microsporidian DNA and bacteria belonging to Spiroplasmataceae and Leuconostocaceae, each represented by a single species, Spiroplasma sp. PL03 and Weissella cf. viridescens, respectively. Additionally, W. cf. viridescens was observed only in microsporidian-infected mosquitoes. More extensive research, including intensive and varied host sampling, as well as determination of metabolic activities based on quantitative methods, should be carried out to confirm our results.
Collapse
Affiliation(s)
- Artur Trzebny
- Molecular Biology Techniques Laboratory, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland.
| | - Anna Slodkowicz-Kowalska
- Department of Biology and Medical Parasitology, Poznan University of Medical Sciences, Poznan, Poland
| | - Johanna Björkroth
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Miroslawa Dabert
- Molecular Biology Techniques Laboratory, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| |
Collapse
|
24
|
Zhao L, Ma YM, Yang B, Han WX, Zhao WH, Chai HL, Zhang ZS, Zhan YJ, Wang LF, Xing Y, Yu LF, Wang JL, Ding YL, Liu YH. Comparative analysis of microbial communities in different growth stages of Dermacentor nuttalli. Front Vet Sci 2022; 9:1021426. [PMID: 36311671 PMCID: PMC9614212 DOI: 10.3389/fvets.2022.1021426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/21/2022] [Indexed: 11/04/2022] Open
Abstract
Ticks were identified as arthropods that are pathogenic vectors. Dermacentor nuttalli is one of the dominant tick species in Inner Mongolia, and it carries and transmits a wide range of pathogenic microorganisms. However, at present, only the detection of D. nuttalli adult ticks and D. nuttalli different developmental stages carrying one specific pathogen, or the next-generation sequencing of D. nuttalli adult ticks were available. In this study, we investigated the microbial community structures of D. nuttalli in different growth stages under laboratory artificial feeding conditions. Total DNA was extracted from seven growth stages (female adult ticks, eggs, larval ticks, engorged larval ticks, nymphal ticks, engorged nymphal ticks, and second-generation adult ticks) obtained from laboratory artificial feeding of engorged D. nuttalli female ticks in Inner Mongolia. Then, the 16S rDNA V3-V4 hypervariable region was amplified to construct an Illumina PE250 library. Finally, 16S rRNA sequencing was performed on Illumina Novaseq 6000 platform. The sequencing data were analyzed using molecular biology software and platforms. The Illumina PE250 sequencing results showed that the egg stage had the highest diversity and number of species (28.74%, 98/341), while the engorged nymph stage had the lowest diversity and number of species (9.72%, 21/216). A total of 387 genera of 22 phyla were annotated in D. nuttalli, with 9 phyla and 57 genera found throughout all 7 growth stages. The dominant phylum was Proteobacteria; the dominant genera were Arsenophonus and Rickettsia; and the genera with the highest relative abundance in the 7 growth stages were Pseudomonas, Paenalcaligenes, Arsenophonus, Arsenophonus, Pseudomonas, Arsenophonus, and Rickettsia, respectively. Among the 23 exact species annotated, Brucella melitensis exhibits pathogeny that poses a serious threat to humans and animals. In this study, the microbial community composition at different growth stages of D. nuttalli was comprehensively analyzed for the first time.
Collapse
Affiliation(s)
- Li Zhao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture and Rural Affairs, Hohhot, China
| | - Yi-Min Ma
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Bo Yang
- Animal Disease Control Center of Ordos, Ordos City, China
| | - Wen-Xiong Han
- Inner Mongolia Saikexing Reproductive Biotechnology (Group) Co., Ltd., Hohhot, China
| | - Wei-Hong Zhao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Hai-Liang Chai
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Zhan-Sheng Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Yong-Jie Zhan
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Li-Feng Wang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
| | - Yu Xing
- Shanghai Origingene Bio-pharm Technology Co. Ltd., Shanghai, China
| | - Lu-Fei Yu
- Shanghai Origingene Bio-pharm Technology Co. Ltd., Shanghai, China
| | - Jin-Ling Wang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture and Rural Affairs, Hohhot, China
| | - Yu-Lin Ding
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture and Rural Affairs, Hohhot, China
| | - Yong-Hong Liu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture and Rural Affairs, Hohhot, China
| |
Collapse
|
25
|
Barillas-Mury C, Ribeiro JMC, Valenzuela JG. Understanding pathogen survival and transmission by arthropod vectors to prevent human disease. Science 2022; 377:eabc2757. [PMID: 36173836 DOI: 10.1126/science.abc2757] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Many endemic poverty-associated diseases, such as malaria and leishmaniasis, are transmitted by arthropod vectors. Pathogens must interact with specific molecules in the vector gut, the microbiota, and the vector immune system to survive and be transmitted. The vertebrate host, in turn, is infected when the pathogen and vector-derived factors, such as salivary proteins, are delivered into the skin by a vector bite. Here, we review recent progress in our understanding of the biology of pathogen transmission from the human to the vector and back, from the vector to the host. We also highlight recent advances in the biology of vector-borne disease transmission, which have translated into additional strategies to prevent human disease by either reducing vector populations or by disrupting their ability to transmit pathogens.
Collapse
Affiliation(s)
- Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | - José M C Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | - Jesus G Valenzuela
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| |
Collapse
|
26
|
Arellano AA, Sommer AJ, Coon KL. Beyond canonical models: why a broader understanding of Diptera-microbiota interactions is essential for vector-borne disease control. Evol Ecol 2022; 37:165-188. [PMID: 37153630 PMCID: PMC10162596 DOI: 10.1007/s10682-022-10197-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Vector-borne diseases constitute a major global public health threat. The most significant arthropod disease vectors are predominantly comprised of members of the insect order Diptera (true flies), which have long been the focus of research into host-pathogen dynamics. Recent studies have revealed the underappreciated diversity and function of dipteran-associated gut microbial communities, with important implications for dipteran physiology, ecology, and pathogen transmission. However, the effective parameterization of these aspects into epidemiological models will require a comprehensive study of microbe-dipteran interactions across vectors and related species. Here, we synthesize recent research into microbial communities associated with major families of dipteran vectors and highlight the importance of development and expansion of experimentally tractable models across Diptera towards understanding the functional roles of the gut microbiota in modulating disease transmission. We then posit why further study of these and other dipteran insects is not only essential to a comprehensive understanding of how to integrate vector-microbiota interactions into existing epidemiological frameworks, but our understanding of the ecology and evolution of animal-microbe symbiosis more broadly.
Collapse
Affiliation(s)
- Aldo A. Arellano
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Andrew J. Sommer
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kerri L. Coon
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
27
|
Karimian F, Koosha M, Choubdar N, Oshaghi MA. Comparative analysis of the gut microbiota of sand fly vectors of zoonotic visceral leishmaniasis (ZVL) in Iran; host-environment interplay shapes diversity. PLoS Negl Trop Dis 2022; 16:e0010609. [PMID: 35853080 PMCID: PMC9337680 DOI: 10.1371/journal.pntd.0010609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/29/2022] [Accepted: 06/26/2022] [Indexed: 11/19/2022] Open
Abstract
The development of Leishmania parasites within sand fly vectors occurs entirely in the insect gut lumen, in the presence of symbiotic and commensal bacteria. The impacts of host species and environment on the gut microbiome are currently poorly understood. We employed MiSeq sequencing of the V3-16S rRNA gene amplicons to characterize and compare the gut microbiota of field-collected populations of Phlebotomus kandelakii, P. perfiliewi, P. alexandri, and P. major, the primary or secondary vectors of zoonotic visceral leishmaniasis (ZVL) in three distinct regions of Iran where ZVL is endemic. In total, 160,550 quality-filtered reads of the V3 region yielded a total of 72 operational taxonomic units (OTUs), belonging to 23 phyla, 47 classes, 91 orders, 131 families, and 335 genera. More than 50% of the bacteria identified were Proteobacteria, followed by Firmicutes (22%), Deinococcus-Thermus (9%), Actinobacteria (6%), and Bacteroidetes (5%). The core microbiome was dominated by eight genera: Acinetobacter, Streptococcus, Enterococcus, Staphylococcus, Bacillus, Propionibacterium, Kocuria, and Corynebacterium. Wolbachia were found in P. alexandri and P. perfiliewi, while Asaia sp. was reported in P. perfiliewi. Substantial variations in the gut bacterial composition were found between geographically distinct populations of the same sand fly species, as well as between different species at the same location, suggesting that sand fly gut microbiota is shaped by both the host species and geographical location. Phlebotomus kandelakii and P. perfiliewi in the northwest, and P. alexandri in the south, the major ZVL vectors, harbor the highest bacterial diversity, suggesting a possible relationship between microbiome diversity and the capacity for parasite transmission. In addition, large numbers of gram-positive human or animal pathogens were found, suggesting that sand fly vectors of ZVL could pose a potential additional threat to livestock and humans in the region studied. The presence of Bacillus subtilis, Enterobacter cloacae, and Asaia sp suggests that these bacteria could be promising candidates for a paratransgenesis approach to the fight against Leishmaniasis. Leishmania infantum, a parasitic protozoan causing fatal visceral leishmaniasis, is transmitted to humans by several sand fly vectors. In this study, the microbiota within the midguts of Phlebotomus kandelakii, P. perfiliewi, P. major and P. alexandri was analyzed by 16S ribosomal DNA (rDNA) Miseq sequencing, revealing highly diverse community composition and abundance, from three diverse ecological and geographical regions of Iran. It appears that the gut microbiota is highly dynamic and controlled by multiple factors, including sand fly host and environment. Proteobacteria were the principal bacterial phylum isolated. High numbers of gram-positive human or animal pathogens were also found, suggesting that sand fly vectors of ZVL could pose a potential threat to livestock and human in the region. Furthermore, there was a positive correlation between vector capacity and bacterial diversities, where the weakest ZVL vector had the lowest diversity, whereas other, more efficient, vectors had higher diversity. This study showed that Bacillus subtilis, Asaia sp. and Enterobacter cloacae are possible candidates for a paratransgenic approach to reduce Leishmania transmission.
Collapse
Affiliation(s)
- Fateh Karimian
- Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
| | - Mona Koosha
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Nayyereh Choubdar
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Oshaghi
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- * E-mail:
| |
Collapse
|
28
|
Omondi ZN, Arserim SK, Töz S, Özbel Y. Host-Parasite Interactions: Regulation of Leishmania Infection in Sand Fly. Acta Parasitol 2022; 67:606-618. [PMID: 35107776 DOI: 10.1007/s11686-022-00519-3] [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: 10/26/2021] [Accepted: 01/11/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE Sand flies are the only proven vectors of leishmaniases, a tropical neglected disease endemic in at least 92 countries. Vector-parasite interactions play a significant role in vector-borne disease transmission. There are various bottlenecks to Leishmania colonization of the sand fly midgut. Such bottlenecks include the production of innate immune-related molecules, digestive proteases, parasite impermeable peritrophic membrane, and resident gut microbiota. These barriers determine the parasite load transmitted and, consequently, the disease outcome in mammalian host. Therefore, it is important to understand the molecular responses of both sand fly and Leishmania during infection. METHOD Here, we reviewed the published literature on sand fly-Leishmania interactions bringing together earlier and current findings to highlight new developments and research gaps in the field. CONCLUSION Recent research studies on sand fly-Leishmania interaction have revealed contrasting observations to past studies. However, how Leishmania parasites evade the sand fly immune response still needs further research. Sand fly response to Leishmania infection can be best understood by analyzing its tissue transcriptome. Better characterization of the role of midgut components could be a game changer in development of transmission-blocking strategies for leishmaniasis.
Collapse
Affiliation(s)
- Zeph Nelson Omondi
- Department of Biology, Faculty of Science, Ege University, Erzene Street, 35040, Bornova/Izmir, Turkey.
| | - Suha Kenan Arserim
- Vocational School of Health Sciences, Manisa Celal Bayar University, Manisa, Turkey
| | - Seray Töz
- Department of Parasitology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Yusuf Özbel
- Department of Parasitology, Faculty of Medicine, Ege University, Izmir, Turkey
| |
Collapse
|
29
|
Metabolic interactions between disease-transmitting vectors and their microbiota. Trends Parasitol 2022; 38:697-708. [DOI: 10.1016/j.pt.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 11/21/2022]
|
30
|
Cecílio P, Cordeiro-da-Silva A, Oliveira F. Sand flies: Basic information on the vectors of leishmaniasis and their interactions with Leishmania parasites. Commun Biol 2022; 5:305. [PMID: 35379881 PMCID: PMC8979968 DOI: 10.1038/s42003-022-03240-z] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/08/2022] [Indexed: 11/09/2022] Open
Abstract
Blood-sucking arthropods transmit a variety of human pathogens acting as disseminators of the so-called vector-borne diseases. Leishmaniasis is a spectrum of diseases caused by different Leishmania species, transmitted quasi worldwide by sand flies. However, whereas many laboratories focus on the disease(s) and etiological agents, considerably less study the respective vectors. In fact, information on sand flies is neither abundant nor easy to find; aspects including basic biology, ecology, and sand-fly-Leishmania interactions are usually reported separately. Here, we compile elemental information on sand flies, in the context of leishmaniasis. We discuss the biology, distribution, and life cycle, the blood-feeding process, and the Leishmania-sand fly interactions that govern parasite transmission. Additionally, we highlight some outstanding questions that need to be answered for the complete understanding of parasite–vector–host interactions in leishmaniasis. In this review, numerous aspects of sand flies as vectors of Leishmania parasites—from biology to the vector parasite interactions—are discussed.
Collapse
Affiliation(s)
- Pedro Cecílio
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA. .,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal. .,Parasite Disease Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal. .,Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto, Portugal.
| | - Anabela Cordeiro-da-Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Parasite Disease Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto, Portugal
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
31
|
Caragata EP, Short SM. Vector microbiota and immunity: modulating arthropod susceptibility to vertebrate pathogens. CURRENT OPINION IN INSECT SCIENCE 2022; 50:100875. [PMID: 35065286 DOI: 10.1016/j.cois.2022.100875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 01/03/2022] [Accepted: 01/13/2022] [Indexed: 06/14/2023]
Abstract
Arthropods, including mosquitoes, sand flies, tsetse flies, and ticks are vectors of many bacterial, parasitic, and viral pathogens that cause serious disease in humans and animals. Their microbiota, that is, all microorganisms that dwell within their tissues, can impact vector immunity and susceptibility to pathogen infection. Historically, host-pathogen-microbiota interactions have not been well described, with little known about mechanism. In this review, we highlight recent advances in understanding how individual microorganisms and microbial communities interact with vectors and human pathogens, the mechanisms they utilize to achieve these effects, and the potential for exploiting these interactions to control pathogen transmission. These studies fill important knowledge gaps and further our understanding of the roles that the vector microbiota plays in pathogen transmission.
Collapse
Affiliation(s)
- Eric P Caragata
- Florida Medical Entomology Laboratory, Department of Entomology and Nematology, Institute of Food and Agricultural Sciences, University of Florida, Vero Beach, FL 32962, USA
| | - Sarah M Short
- Department of Entomology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
32
|
Eberhard FE, Klimpel S, Guarneri AA, Tobias NJ. Exposure to Trypanosoma parasites induces changes in the microbiome of the Chagas disease vector Rhodnius prolixus. MICROBIOME 2022; 10:45. [PMID: 35272716 PMCID: PMC8908696 DOI: 10.1186/s40168-022-01240-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/31/2022] [Indexed: 05/04/2023]
Abstract
BACKGROUND The causative agent of Chagas disease, Trypanosoma cruzi, and its nonpathogenic relative, Trypanosoma rangeli, are transmitted by haematophagous triatomines and undergo a crucial ontogenetic phase in the insect's intestine. In the process, the parasites interfere with the host immune system as well as the microbiome present in the digestive tract potentially establishing an environment advantageous for development. However, the coherent interactions between host, pathogen and microbiota have not yet been elucidated in detail. We applied a metagenome shotgun sequencing approach to study the alterations in the microbiota of Rhodnius prolixus, a major vector of Chagas disease, after exposure to T. cruzi and T. rangeli focusing also on the functional capacities present in the intestinal microbiome of the insect. RESULTS The intestinal microbiota of R. prolixus was dominated by the bacterial orders Enterobacterales, Corynebacteriales, Lactobacillales, Clostridiales and Chlamydiales, whereas the latter conceivably originated from the blood used for pathogen exposure. The anterior and posterior midgut samples of the exposed insects showed a reduced overall number of organisms compared to the control group. However, we also found enriched bacterial groups after exposure to T. cruzi as well as T rangeli. While the relative abundance of Enterobacterales and Corynebacteriales decreased considerably, the Lactobacillales, mainly composed of the genus Enterococcus, developed as the most abundant taxonomic group. This applies in particular to vectors challenged with T. rangeli and at early timepoints after exposure to vectors challenged with T. cruzi. Furthermore, we were able to reconstruct four metagenome-assembled genomes from the intestinal samples and elucidate their unique metabolic functionalities within the triatomine microbiome, including the genome of a recently described insect symbiont, Candidatus Symbiopectobacterium, and the secondary metabolites producing bacteria Kocuria spp. CONCLUSIONS Our results facilitate a deeper understanding of the processes that take place in the intestinal tract of triatomine vectors during colonisation by trypanosomal parasites and highlight the influential aspects of pathogen-microbiota interactions. In particular, the mostly unexplored metabolic capacities of the insect vector's microbiome are clearer, underlining its role in the transmission of Chagas disease. Video Abstract.
Collapse
Affiliation(s)
- Fanny E. Eberhard
- Institute for Ecology, Evolution and Diversity, Goethe University Frankfurt, Biologicum Campus Riedberg, Max-von-Laue-Str. 13, 60439 Frankfurt/Main, Germany
| | - Sven Klimpel
- Institute for Ecology, Evolution and Diversity, Goethe University Frankfurt, Biologicum Campus Riedberg, Max-von-Laue-Str. 13, 60439 Frankfurt/Main, Germany
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE TBG), Senckenberganlage 25, 60325 Frankfurt/Main, Germany
- Senckenberg Gesellschaft für Naturforschung, Senckenberg Biodiversity and Climate Research Centre, Senckenberganlage 25, 60325 Frankfurt/Main, Germany
| | - Alessandra A. Guarneri
- Vector Behaviour and Pathogen Interaction Group, Instituto René Rachou, Avenida Augusto de Lima,1715, Belo Horizonte, MG CEP 30190-009 Brazil
| | - Nicholas J. Tobias
- LOEWE Centre for Translational Biodiversity Genomics (LOEWE TBG), Senckenberganlage 25, 60325 Frankfurt/Main, Germany
- Senckenberg Gesellschaft für Naturforschung, Senckenberg Biodiversity and Climate Research Centre, Senckenberganlage 25, 60325 Frankfurt/Main, Germany
| |
Collapse
|
33
|
Telleria EL, Azevedo-Brito DA, Kykalová B, Tinoco-Nunes B, Pitaluga AN, Volf P, Traub-Csekö YM. Leishmania infantum Infection Modulates the Jak-STAT Pathway in Lutzomyia longipalpis LL5 Embryonic Cells and Adult Females, and Affects Parasite Growth in the Sand Fly. FRONTIERS IN TROPICAL DISEASES 2021. [DOI: 10.3389/fitd.2021.747820] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Phlebotomine sand flies (Diptera, Psychodidae) belonging to the Lutzomyia genus transmit zoonoses in the New World. Lutzomyia longipalpis is the main vector of Leishmania infantum, which is the causative agent of visceral leishmaniasis in Brazil. To identify key molecular aspects involved in the interaction between vector and pathogens and contribute to developing disease transmission controls, we investigated the sand fly innate immunity mediated by the Janus kinase/signal transducer and activator of transcription (Jak-STAT) pathway in response to L. infantum infection. We used two study models: L. longipalpis LL5 embryonic cells co-cultured with L. infantum and sand fly females artificially infected with the parasite. We used qPCR to follow the L. longipalpis gene expression of molecules involved in the Jak-STAT pathway. Also, we modulated the Jak-STAT mediated immune response to understand its role in Leishmania parasite infection. For that, we used RNAi to silence the pathway regulators, protein inhibitor of activated STATs (PIAS) in LL5 cells, and STAT in adult females. In addition, the pathway suppression effect on parasite development within the vector was assessed by light microscopy in late-phase infection. The silencing of the repressor PIAS in LL5 cells led to a moderate increase in a protein tyrosine phosphatase 61F (PTP61F) expression. It suggests a compensatory regulation between these two repressors. L. infantum co-culture with LL5 cells upregulated repressors PIAS, suppressor of cytokine signaling (SOCS), and PTP61F. It also downmodulated virus-induced RNA-1 (VIR-1), a pathway effector, indicating that the parasite could repress the Jak-STAT pathway in LL5 cells. In Leishmania-infected L. longipalpis females, STAT and the antimicrobial peptide attacin were downregulated on the third day post-infection, suggesting a correlation that favors the parasite survival at the end of blood digestion in the sand fly. The antibiotic treatment of infected females showed that the reduction of gut bacteria had little effect on the Jak-STAT pathway regulation. STAT gene silencing mediated by RNAi reduced the expression of inducible nitric oxide synthase (iNOS) and favored Leishmania growth in sand flies on the first day post-infection. These results indicate that STAT participated in the iNOS regulation with subsequent effect on parasite survival.
Collapse
|
34
|
Microbiomes of Blood-Feeding Arthropods: Genes Coding for Essential Nutrients and Relation to Vector Fitness and Pathogenic Infections. A Review. Microorganisms 2021; 9:microorganisms9122433. [PMID: 34946034 PMCID: PMC8704530 DOI: 10.3390/microorganisms9122433] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 11/08/2021] [Accepted: 11/20/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Blood-feeding arthropods support a diverse array of symbiotic microbes, some of which facilitate host growth and development whereas others are detrimental to vector-borne pathogens. We found a common core constituency among the microbiota of 16 different arthropod blood-sucking disease vectors, including Bacillaceae, Rickettsiaceae, Anaplasmataceae, Sphingomonadaceae, Enterobacteriaceae, Pseudomonadaceae, Moraxellaceae and Staphylococcaceae. By comparing 21 genomes of common bacterial symbionts in blood-feeding vectors versus non-blooding insects, we found that certain enteric bacteria benefit their hosts by upregulating numerous genes coding for essential nutrients. Bacteria of blood-sucking vectors expressed significantly more genes (p < 0.001) coding for these essential nutrients than those of non-blooding insects. Moreover, compared to endosymbionts, the genomes of enteric bacteria also contained significantly more genes (p < 0.001) that code for the synthesis of essential amino acids and proteins that detoxify reactive oxygen species. In contrast, microbes in non-blood-feeding insects expressed few gene families coding for these nutrient categories. We also discuss specific midgut bacteria essential for the normal development of pathogens (e.g., Leishmania) versus others that were detrimental (e.g., bacterial toxins in mosquitoes lethal to Plasmodium spp.).
Collapse
|
35
|
Kykalová B, Tichá L, Volf P, Loza Telleria E. Phlebotomus papatasi Antimicrobial Peptides in Larvae and Females and a Gut-Specific Defensin Upregulated by Leishmania major Infection. Microorganisms 2021; 9:microorganisms9112307. [PMID: 34835433 PMCID: PMC8625375 DOI: 10.3390/microorganisms9112307] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 12/30/2022] Open
Abstract
Phlebotomus papatasi is the vector of Leishmania major, causing cutaneous leishmaniasis in the Old World. We investigated whether P. papatasi immunity genes were expressed toward L. major, commensal gut microbes, or a combination of both. We focused on sand fly transcription factors dorsal and relish and antimicrobial peptides (AMPs) attacin and defensin and assessed their relative gene expression by qPCR. Sand fly larvae were fed food with different bacterial loads. Relish and AMPs gene expressions were higher in L3 and early L4 larval instars, while bacteria 16S rRNA increased in late L4 larval instar, all fed rich-microbe food compared to the control group fed autoclaved food. Sand fly females were treated with an antibiotic cocktail to deplete gut bacteria and were experimentally infected by Leishmania. Compared to non-infected females, dorsal and defensin were upregulated at early and late infection stages, respectively. An earlier increase of defensin was observed in infected females when bacteria recolonized the gut after the removal of antibiotics. Interestingly, this defensin gene expression occurred specifically in midguts but not in other tissues of females and larvae. A gut-specific defensin gene upregulated by L. major infection, in combination with gut-bacteria, is a promising molecular target for parasite control strategies.
Collapse
|
36
|
Yang J, Yu P, Liu X, Zhao J, Zhang H, Chen W. Shifts in diversity and function of bacterial community during manufacture of Rushan. J Dairy Sci 2021; 104:12375-12393. [PMID: 34482971 DOI: 10.3168/jds.2021-20654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/19/2021] [Indexed: 01/04/2023]
Abstract
Rushan is a traditional dairy product consumed by the Bai people in the Yunnan Province of China, and its production still follows the traditional procedure of backslopping. However, how the microbial composition of raw materials and processing shape the microorganisms in Rushan have not been systemically reported. In this study, high-throughput sequencing technique was applied to analyze the microbial compositions of raw milk, fresh Rushan, curd whey, acid whey, and dry Rushan at the phylum, family, genus, and Lactobacillus species levels. The results indicated that Lactobacillus, Lactococcus, and Streptococcus were dominant genera in Rushan, whereas Lactobacillus kefiranofaciens and Lactobacillus helveticus were the 2 abundant species at the Lactobacillus species level. The network analysis indicated that raw milk mainly contributed to the microbial diversity of Rushan, whereas acid whey made a great contribution to shaping the relative abundance of microbes in Rushan and dramatically increased acid-producing genera, such as Lactobacillus and Acetobacter. The variation in microbial composition led to an increase in the relative abundance of pathways related to energy supply, acid production, fatty acid accumulation, cysteine, methionine, and lysine accumulation. The volatile profile of Rushan was rich in esters and acids, and the high relative abundance of Lactobacillus might be associated with reduction of amino acid metabolism, degradation of unpleasant flavored xylene, and accumulation of decanoic, dodecanoic, and tetradecanoic acids in the products. The accumulation of medium long-chain fatty acids might result from the relative abundance of FabF, FabZ, and FabI, particularly from Lactobacillus amylolyticus and Lacticaseibacillus paracasei.
Collapse
Affiliation(s)
- Jiang Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Peng Yu
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xiaoming Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; International Joint Research Laboratory for Pharmabiotics and Antibiotic Resistance, Jiangnan University, Wuxi, Jiangsu 214122, China.
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China; National Engineering Research Centre for Functional Food, Wuxi, Jiangsu 214122, China
| |
Collapse
|
37
|
Balaska S, Fotakis EA, Chaskopoulou A, Vontas J. Chemical control and insecticide resistance status of sand fly vectors worldwide. PLoS Negl Trop Dis 2021; 15:e0009586. [PMID: 34383751 PMCID: PMC8360369 DOI: 10.1371/journal.pntd.0009586] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Phlebotomine sand flies are prominent vectors of Leishmania parasites that cause leishmaniasis, which comes second to malaria in terms of parasitic causative fatalities globally. In the absence of human vaccines, sand fly chemical-based vector control is a key component of leishmaniasis control efforts. METHODS AND FINDINGS We performed a literature review on the current interventions, primarily, insecticide-based used for sand fly control, as well as the global insecticide resistance (IR) status of the main sand fly vector species. Indoor insecticidal interventions, such as residual spraying and treated bed nets are the most widely deployed, while several alternative control strategies are also used in certain settings and/or are under evaluation. IR has been sporadically detected in sand flies in India and other regions, using non-standardized diagnostic bioassays. Molecular studies are limited to monitoring of known pyrethroid resistance mutations (kdr), which are present at high frequencies in certain regions. CONCLUSIONS As the leishmaniasis burden remains a major problem at a global scale, evidence-based rational use of insecticidal interventions is required to meet public health demands. Standardized bioassays and molecular markers are a prerequisite for this task, albeit are lagging behind. Experiences from other disease vectors underscore the need for the implementation of appropriate IR management (IRM) programs, in the framework of integrated vector management (IVM). The implementation of alternative strategies seems context- and case-specific, with key eco-epidemiological parameters yet to be investigated. New biotechnology-based control approaches might also come into play in the near future to further reinforce sand fly/leishmaniasis control efforts.
Collapse
Affiliation(s)
- Sofia Balaska
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, Heraklion, Greece
- Department of Biology, University of Crete, Heraklion, Greece
| | - Emmanouil Alexandros Fotakis
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, Heraklion, Greece
- Department of Crop Science, Agricultural University of Athens, Athens, Greece
| | | | - John Vontas
- Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology Hellas, Heraklion, Greece
- Department of Crop Science, Agricultural University of Athens, Athens, Greece
- * E-mail:
| |
Collapse
|
38
|
Leishmaniasis: the act of transmission. Trends Parasitol 2021; 37:976-987. [PMID: 34389215 DOI: 10.1016/j.pt.2021.07.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 02/06/2023]
Abstract
The contribution of vector transmission to pathogen establishment is largely underrated. For Leishmania, transmission by sand flies is critical to early survival involving an irreproducible myriad of parasite, vector, and host molecules acting in concert to promote infection at the bite site. Here, we review recent breakthroughs that provide consequential insights into how vector transmission of Leishmania unfolds. We focus on recent work pertaining to the effect of gut microbiota, sand fly immunity, and changes in metacyclogenesis upon multiple blood meals, on Leishmania development and transmission. We also explore how sand fly saliva, egested parasite molecules and vector gut microbiota, and bleeding have been implicated in modulating the early innate host response to Leishmania, affecting the phenotype of neutrophils and monocytes arriving at the bite site.
Collapse
|
39
|
Vasconcelos EJR, Roy C, Geiger JA, Oney KM, Koo M, Ren S, Oakley BB, Diniz PPVP. Data analysis workflow for the detection of canine vector-borne pathogens using 16 S rRNA Next-Generation Sequencing. BMC Vet Res 2021; 17:262. [PMID: 34332568 PMCID: PMC8325813 DOI: 10.1186/s12917-021-02969-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 07/16/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Vector-borne diseases (VBDs) impact both human and veterinary medicine and pose special public health challenges. The main bacterial vector-borne pathogens (VBPs) of importance in veterinary medicine include Anaplasma spp., Bartonella spp., Ehrlichia spp., and Spotted Fever Group Rickettsia. Taxon-targeted PCR assays are the current gold standard for VBP diagnostics but limitations on the detection of genetically diverse organisms support a novel approach for broader detection of VBPs. We present a methodology for genetic characterization of VBPs using Next-Generation Sequencing (NGS) and computational approaches. A major advantage of NGS is the ability to detect multiple organisms present in the same clinical sample in an unsupervised (i.e. non-targeted) and semi-quantitative way. The Standard Operating Procedure (SOP) presented here combines industry-standard microbiome analysis tools with our ad-hoc bioinformatic scripts to form a complete analysis pipeline accessible to veterinary scientists and freely available for download and use at https://github.com/eltonjrv/microbiome.westernu/tree/SOP . RESULTS We tested and validated our SOP by mimicking single, double, and triple infections in genomic canine DNA using serial dilutions of plasmids containing the entire 16 S rRNA gene sequence of (A) phagocytophilum, (B) v. berkhoffii, and E. canis. NGS with broad-range 16 S rRNA primers followed by our bioinformatics SOP was capable of detecting these pathogens in biological replicates of different dilutions. These results illustrate the ability of NGS to detect and genetically characterize multi-infections with different amounts of pathogens in a single sample. CONCLUSIONS Bloodborne microbiomics & metagenomics approaches may help expand the molecular diagnostic toolbox in veterinary and human medicine. In this paper, we present both in vitro and in silico detailed protocols that can be combined into a single workflow that may provide a significant improvement in VBP diagnostics and also facilitate future applications of microbiome research in veterinary medicine.
Collapse
Affiliation(s)
- Elton J. R. Vasconcelos
- College of Veterinary Medicine, Western University of Health Sciences, 309 East 2nd Street, CA 91766 − 1854 Pomona, USA
- Leeds Omics, University of Leeds, LS2 9JT Leeds, United Kingdom
| | - Chayan Roy
- College of Veterinary Medicine, Western University of Health Sciences, 309 East 2nd Street, CA 91766 − 1854 Pomona, USA
| | - Joseph A. Geiger
- College of Veterinary Medicine, Western University of Health Sciences, 309 East 2nd Street, CA 91766 − 1854 Pomona, USA
| | - Kristina M. Oney
- College of Veterinary Medicine, Western University of Health Sciences, 309 East 2nd Street, CA 91766 − 1854 Pomona, USA
| | - Melody Koo
- College of Veterinary Medicine, Western University of Health Sciences, 309 East 2nd Street, CA 91766 − 1854 Pomona, USA
| | - Songyang Ren
- College of Veterinary Medicine, Western University of Health Sciences, 309 East 2nd Street, CA 91766 − 1854 Pomona, USA
| | - Brian B. Oakley
- College of Veterinary Medicine, Western University of Health Sciences, 309 East 2nd Street, CA 91766 − 1854 Pomona, USA
| | - Pedro Paulo V. P. Diniz
- College of Veterinary Medicine, Western University of Health Sciences, 309 East 2nd Street, CA 91766 − 1854 Pomona, USA
| |
Collapse
|
40
|
Lutzomyia longipalpis Antimicrobial Peptides: Differential Expression during Development and Potential Involvement in Vector Interaction with Microbiota and Leishmania. Microorganisms 2021; 9:microorganisms9061271. [PMID: 34207941 PMCID: PMC8230673 DOI: 10.3390/microorganisms9061271] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/04/2021] [Accepted: 02/15/2021] [Indexed: 12/25/2022] Open
Abstract
Antimicrobial peptides (AMPs) are produced to control bacteria, fungi, protozoa, and other infectious agents. Sand fly larvae develop and feed on a microbe-rich substrate, and the hematophagous females are exposed to additional pathogens. We focused on understanding the role of the AMPs attacin (Att), cecropin (Cec), and four defensins (Def1, Def2, Def3, and Def4) in Lutzomyia longipalpis, the main vector of visceral leishmaniasis in the Americas. Larvae and adults were collected under different feeding regimens, in addition to females artificially infected by Leishmania infantum. AMPs’ gene expression was assessed by qPCR, and gene function of Att and Def2 was investigated by gene silencing. The gene knockdown effect on bacteria and parasite abundance was evaluated by qPCR, and parasite development was verified by light microscopy. We demonstrate that L. longipalpis larvae and adults trigger AMPs expression during feeding, which corresponds to an abundant presence of bacteria. Att and Def2 expression were significantly increased in Leishmania-infected females, while Att suppression favored bacteria growth. In conclusion, L. longipalpis AMPs’ expression is tuned in response to bacteria and parasites but does not seem to interfere with the Leishmania cycle.
Collapse
|
41
|
Gut Microbiota Dynamics in Natural Populations of Pintomyia evansi under Experimental Infection with Leishmania infantum. Microorganisms 2021; 9:microorganisms9061214. [PMID: 34199688 PMCID: PMC8228094 DOI: 10.3390/microorganisms9061214] [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: 02/23/2021] [Revised: 05/06/2021] [Accepted: 05/19/2021] [Indexed: 01/04/2023] Open
Abstract
Pintomyia evansi is recognized by its vectorial competence in the transmission of parasites that cause fatal visceral leishmaniasis in rural and urban environments of the Caribbean coast of Colombia. The effect on and the variation of the gut microbiota in female P. evansi infected with Leishmania infantum were evaluated under experimental conditions using 16S rRNA Illumina MiSeq sequencing. In the coinfection assay with L. infantum, 96.8% of the midgut microbial population was composed mainly of Proteobacteria (71.0%), followed by Cyanobacteria (20.4%), Actinobacteria (2.7%), and Firmicutes (2.7%). In insect controls (uninfected with L. infantum) that were treated or not with antibiotics, Ralstonia was reported to have high relative abundance (55.1–64.8%), in contrast to guts with a high load of infection from L. infantum (23.4–35.9%). ASVs that moderately increased in guts infected with Leishmania were Bacillus and Aeromonas. Kruskal–Wallis nonparametric variance statistical inference showed statistically significant intergroup differences in the guts of P. evansi infected and uninfected with L. infantum (p < 0.05), suggesting that some individuals of the microbiota could induce or restrict Leishmania infection. This assay also showed a negative effect of the antibiotic treatment and L. infantum infection on the gut microbiota diversity. Endosymbionts, such as Microsporidia infections (<2%), were more often associated with guts without Leishmania infection, whereas Arsenophonus was only found in guts with a high load of Leishmania infection and treated with antibiotics. Finally, this is the first report that showed the potential role of intestinal microbiota in natural populations of P. evansi in susceptibility to L. infantum infection.
Collapse
|
42
|
Zayats R, Uzonna JE, Murooka TT. Visualizing the In Vivo Dynamics of Anti- Leishmania Immunity: Discoveries and Challenges. Front Immunol 2021; 12:671582. [PMID: 34093571 PMCID: PMC8172142 DOI: 10.3389/fimmu.2021.671582] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/07/2021] [Indexed: 11/20/2022] Open
Abstract
Intravital microscopy, such as 2-photon microscopy, is now a mainstay in immunological research to visually characterize immune cell dynamics during homeostasis and pathogen infections. This approach has been especially beneficial in describing the complex process of host immune responses to parasitic infections in vivo, such as Leishmania. Human-parasite co-evolution has endowed parasites with multiple strategies to subvert host immunity in order to establish chronic infections and ensure human-to-human transmission. While much focus has been placed on viral and bacterial infections, intravital microscopy studies during parasitic infections have been comparatively sparse. In this review, we will discuss how in vivo microscopy has provided important insights into the generation of innate and adaptive immunity in various organs during parasitic infections, with a primary focus on Leishmania. We highlight how microscopy-based approaches may be key to providing mechanistic insights into Leishmania persistence in vivo and to devise strategies for better parasite control.
Collapse
Affiliation(s)
- Romaniya Zayats
- Rady Faculty of Health Sciences, Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Jude E. Uzonna
- Rady Faculty of Health Sciences, Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
- Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| | - Thomas T. Murooka
- Rady Faculty of Health Sciences, Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
- Rady Faculty of Health Sciences, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
43
|
Rêgo FD, Soares RP. Lutzomyia longipalpis: an update on this sand fly vector. AN ACAD BRAS CIENC 2021; 93:e20200254. [PMID: 33950136 DOI: 10.1590/0001-37652021xxxx] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/17/2020] [Indexed: 12/13/2022] Open
Abstract
Lutzomyia longipalpis is the most important vector of Leishmania infantum, the etiological agent of visceral leishmaniasis (VL) in the New World. It is a permissive vector susceptible to infection with several Leishmania species. One of the advantages that favors the study of this sand fly is the possibility of colonization in the laboratory. For this reason, several researchers around the world use this species as a model for different subjects including biology, insecticides testing, host-parasite interaction, physiology, genetics, proteomics, molecular biology, and saliva among others. In 2003, we published our first review (Soares & Turco 2003) on this vector covering several aspects of Lu. longipalpis. This current review summarizes what has been published between 2003-2020. During this period, modern approaches were incorporated following the development of more advanced and sensitive techniques to assess this sand fly.
Collapse
Affiliation(s)
- Felipe D Rêgo
- Fundação Oswaldo Cruz (FIOCRUZ/MG), Instituto René Rachou, Avenida Augusto de Lima, 1715, Barro Preto, 30180-104 Belo Horizonte, MG, Brazil
| | - Rodrigo Pedro Soares
- Fundação Oswaldo Cruz (FIOCRUZ/MG), Instituto René Rachou, Avenida Augusto de Lima, 1715, Barro Preto, 30180-104 Belo Horizonte, MG, Brazil
| |
Collapse
|
44
|
Gabrieli P, Caccia S, Varotto-Boccazzi I, Arnoldi I, Barbieri G, Comandatore F, Epis S. Mosquito Trilogy: Microbiota, Immunity and Pathogens, and Their Implications for the Control of Disease Transmission. Front Microbiol 2021; 12:630438. [PMID: 33889137 PMCID: PMC8056039 DOI: 10.3389/fmicb.2021.630438] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/02/2021] [Indexed: 11/16/2022] Open
Abstract
In mosquitoes, the interaction between the gut microbiota, the immune system, and the pathogens that these insects transmit to humans and animals is regarded as a key component toward the development of control strategies, aimed at reducing the burden of severe diseases, such as malaria and dengue fever. Indeed, different microorganisms from the mosquito microbiota have been investigated for their ability to affect important traits of the biology of the host insect, related with its survival, development and reproduction. Furthermore, some microorganisms have been shown to modulate the immune response of mosquito females, significantly shaping their vector competence. Here, we will review current knowledge in this field, focusing on i) the complex interaction between the intestinal microbiota and mosquito females defenses, both in the gut and at humoral level; ii) how knowledge on these issues contributes to the development of novel and targeted strategies for the control of mosquito-borne diseases such as the use of paratransgenesis or taking advantage of the relationship between Wolbachia and mosquito hosts. We conclude by providing a brief overview of available knowledge on microbiota-immune system interplay in major insect vectors.
Collapse
Affiliation(s)
- Paolo Gabrieli
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | - Silvia Caccia
- Department of Agricultural Sciences, University of Naples "Federico II", Naples, Italy.,Task Force on Microbiome Studies, University of Naples "Federico II", Naples, Italy
| | - Ilaria Varotto-Boccazzi
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | - Irene Arnoldi
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Giulia Barbieri
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Francesco Comandatore
- "L. Sacco" Department of Biomedical and Clinical Sciences, Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | - Sara Epis
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| |
Collapse
|
45
|
Garlapati R, Iniguez E, Serafim TD, Mishra PK, Rooj B, Sinha B, Valenzuela JG, Srikantiah S, Bern C, Kamhawi S. Towards a Sustainable Vector-Control Strategy in the Post Kala-Azar Elimination Era. Front Cell Infect Microbiol 2021; 11:641632. [PMID: 33768013 PMCID: PMC7985538 DOI: 10.3389/fcimb.2021.641632] [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: 12/14/2020] [Accepted: 02/03/2021] [Indexed: 11/26/2022] Open
Abstract
Visceral leishmaniasis (VL) is a potentially deadly parasitic disease. In the Indian sub-continent, VL is caused by Leishmania donovani and transmitted via the bite of an infected Phlebotomus argentipes female sand fly, the only competent vector species in the region. The highest disease burden is in the northern part of the Indian sub-continent, especially in the state of Bihar. India, Bangladesh, and Nepal embarked on an initiative, coordinated by World Health Organization, to eliminate VL as a public health problem by the year 2020. The main goal is to reduce VL incidence below one case per 10,000 people through early case-detection, prompt diagnosis and treatment, and reduction of transmission using vector control measures. Indoor residual spraying, a major pillar of the elimination program, is the only vector control strategy used by the government of India. Though India is close to its VL elimination target, important aspects of vector bionomics and sand fly transmission dynamics are yet to be determined. To achieve sustained elimination and to prevent a resurgence of VL, knowledge gaps in vector biology and behavior, and the constraints they may pose to current vector control methods, need to be addressed. Herein, we discuss the successes and failures of previous and current vector-control strategies implemented to combat kala-azar in Bihar, India, and identify gaps in our understanding of vector transmission towards development of innovative tools to ensure sustained vector control in the post-elimination period.
Collapse
Affiliation(s)
- Rajesh Garlapati
- Bihar Technical Support Program, CARE India Solutions for Sustainable Development, Patna, India
| | - Eva Iniguez
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Tiago D Serafim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Prabhas K Mishra
- Bihar Technical Support Program, CARE India Solutions for Sustainable Development, Patna, India
| | - Basab Rooj
- Bihar Technical Support Program, CARE India Solutions for Sustainable Development, Patna, India
| | - Bikas Sinha
- Bihar Technical Support Program, CARE India Solutions for Sustainable Development, Patna, India
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Sridhar Srikantiah
- Bihar Technical Support Program, CARE India Solutions for Sustainable Development, Patna, India
| | - Caryn Bern
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, United States
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
46
|
Immune response dynamics and Lutzomyia longipalpis exposure characterize a biosignature of visceral leishmaniasis susceptibility in a canine cohort. PLoS Negl Trop Dis 2021; 15:e0009137. [PMID: 33617528 PMCID: PMC7943000 DOI: 10.1371/journal.pntd.0009137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 03/09/2021] [Accepted: 01/12/2021] [Indexed: 12/29/2022] Open
Abstract
Background Reports have shown correlations between the immune response to vector saliva and Leishmaniasis outcome. We followed dogs in an endemic area for two years characterizing resistance or susceptibility to canine visceral leishmaniasis (CVL) according to Leishmania infantum diagnosis and clinical development criteria. Then, we aimed to identify a biosignature based on parasite load, serum biological mediators’ interactions, and vector exposure intensity associated with CVL resistance and susceptibility. Methodology/Principal findings A prospective two-year study was conducted in an area endemic for CVL. Dogs were evaluated at 6-month intervals to determine infection, clinical manifestations, immune profile, and sandfly exposure. CVL resistance or susceptibility was determined upon the conclusion of the study. After two years, 78% of the dogs were infected with L. infantum (53% susceptible and 47% resistant to CVL). Susceptible dogs presented higher splenic parasite load as well as persistence of the parasite during the follow-up, compared to resistant ones. Susceptible dogs also displayed a higher number of correlations among the investigated biological mediators, before and after infection diagnosis. At baseline, anti-saliva antibodies, indicative of exposure to the vector, were detected in 62% of the dogs, reaching 100% in one year. Higher sandfly exposure increased the risk of susceptibility to CVL by 1.6 times (CI: 1.11–2.41). We identified a discriminatory biosignature between the resistant and susceptible dogs assessing splenic parasite load, interaction of biological mediators, PGE2 serum levels and intensity of exposure to sandfly. All these parameters were elevated in susceptible dogs compared to resistant animals. Conclusions/Significance The biosignature identified in our study reinforces the idea that CVL is a complex multifactorial disease that is affected by a set of factors which are correlated and, for a better understanding of CVL, should not be evaluated in an isolated way. Visceral Leishmaniasis (VL) is a disease that can affect humans and dogs, caused by a parasite called Leishmania transmitted through the bite of sandfly insects. During the bite, together with the parasite, the insects also inoculate their saliva into the host. The host immune response produces molecules to the sandfly saliva, such as antibodies and cytokines that can impact VL resistance or susceptibility. The presence of these molecules also indicates if the insects bit the hosts. We followed dogs of a VL endemic area for two years to study Canine Visceral Leishmaniasis (CVL) and immune response to sandfly saliva. Dogs were evaluated at 6-month intervals to determine Leishmania infection, clinical manifestations, parasite load, immune response, and sandfly exposure. CVL resistance or susceptibility was determined upon the conclusion of the study. Dogs living in the endemic area were intensely bitten, as at the beginning of the study, 62% of the dogs present anti-saliva antibodies, reaching 100% after one year. Our findings revealed a biosignature of CVL susceptibility characterized by elevated parasite load, interaction of cytokines, and higher exposure to the sandfly. This data reinforced that CVL is a complex disease affected by several factors related to each other.
Collapse
|
47
|
Aguiar Martins K, Meirelles MHDA, Mota TF, Abbasi I, de Queiroz ATL, Brodskyn CI, Veras PST, Mothé Fraga DB, Warburg A. Effects of larval rearing substrates on some life-table parameters of Lutzomyia longipalpis sand flies. PLoS Negl Trop Dis 2021; 15:e0009034. [PMID: 33476330 PMCID: PMC7870073 DOI: 10.1371/journal.pntd.0009034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 02/08/2021] [Accepted: 12/03/2020] [Indexed: 11/18/2022] Open
Abstract
Sand flies are the insects responsible for transmitting Leishmania parasites, the causative agents of leishmaniasis in humans. However, the effects of sand fly breeding sites on their biology and ecology remain poorly understood. Herein, we studied how larval nutrition associated with putative breeding sites of the sand fly Lutzomyia longipalpis affects their oviposition, development, microbiome, and susceptibility to Leishmania by rearing L. longipalpis on substrates collected from an endemic area for leishmaniasis in Brazil. The results showed that female L. longipalpis select the oviposition site based on its potential to promote larval maturation and while composting cashew leaf litter hindered the development, larvae reared on chicken feces developed rapidly. Typical gut microbial profiles were found in larvae reared upon cashew leaf litter. Adult females from larvae reared on substrate collected in chicken coops were infected with Leishmania infantum, indicating that they were highly susceptible to the parasite. In conclusion, the larval breeding sites can exert an important role in the epidemiology of leishmaniasis. Sand flies are the insect vectors involved in the transmission of many pathogens, however, the transmission of parasites to humans leading to visceral leishmaniasis is currently the most critical threat caused by this insect. Despite the importance of the vector, many aspects of the biology of sand flies are poorly understood, especially their breeding sites. This study was designed to evaluate the oviposition, life span, microbiome, and parasite infections in the main species of sand fly responsible for visceral leishmaniasis in America. Insects were reared on substrates collected from different putative habitats of sand flies in an endemic area for the disease in Brazil. The results showed that female vectors selected an oviposition site depending on the potential offered to their offspring. Furthermore, the development of immature stages varied according to the type of substrate evaluated, with cashew leaves litter delaying larval development, while chicken shelter promoted larval development. The challenge of females emerging from chicken shelter substrate with the parasite indicates that insects reared in such an environment could successfully sustain the infection. These results suggest that the type of breeding site can affect insect biology as well as the epidemiology of the disease.
Collapse
Affiliation(s)
| | - Maria Helena de Athayde Meirelles
- Instituto Gonçalo Moniz-Fundação Oswaldo Cruz, Salvador, Brazil
- Escola de Medicina Veterinária e Zootecnia-Universidade Federal da Bahia, Salvador, Brazil
| | | | - Ibrahim Abbasi
- Kuvin Center for the Study of Infectious & Tropical Diseases, Department of Microbiology and Molecular Genetics, Institute of Medical Research, Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Claudia Ida Brodskyn
- Instituto Gonçalo Moniz-Fundação Oswaldo Cruz, Salvador, Brazil
- Escola de Medicina Veterinária e Zootecnia-Universidade Federal da Bahia, Salvador, Brazil
| | | | - Deborah Bittencourt Mothé Fraga
- Instituto Gonçalo Moniz-Fundação Oswaldo Cruz, Salvador, Brazil
- Escola de Medicina Veterinária e Zootecnia-Universidade Federal da Bahia, Salvador, Brazil
| | - Alon Warburg
- Kuvin Center for the Study of Infectious & Tropical Diseases, Department of Microbiology and Molecular Genetics, Institute of Medical Research, Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
48
|
Understanding the immune responses involved in mediating protection or immunopathology during leishmaniasis. Biochem Soc Trans 2021; 49:297-311. [PMID: 33449103 DOI: 10.1042/bst20200606] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 01/21/2023]
Abstract
Leishmaniasis is a vector-borne Neglected Tropical Disease (NTD) transmitted by the sand fly and is a major public health problem worldwide. Infections caused by Leishmania clinically manifest as a wide range of diseases, such as cutaneous (CL), diffuse cutaneous (DCL), mucosal (MCL) and visceral leishmaniasis (VL). The host innate and adaptative immune responses play critical roles in the defense against leishmaniasis. However, Leishmania parasites also manipulate the host immune response for their survival and replication. In addition, other factors such as sand fly salivary proteins and microbiota also promote disease susceptibility and parasite spread by modulating local immune response. Thus, a complex interplay between parasite, sand fly and the host immunity governs disease severity and outcome. In this review, we discuss the host immune response during Leishmania infection and highlight the factors associated with resistance or susceptibility.
Collapse
|
49
|
Sloan MA, Sadlova J, Lestinova T, Sanders MJ, Cotton JA, Volf P, Ligoxygakis P. The Phlebotomus papatasi systemic transcriptional response to trypanosomatid-contaminated blood does not differ from the non-infected blood meal. Parasit Vectors 2021; 14:15. [PMID: 33407867 PMCID: PMC7789365 DOI: 10.1186/s13071-020-04498-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/23/2020] [Indexed: 02/13/2023] Open
Abstract
Background Leishmaniasis, caused by parasites of the genus Leishmania, is a disease that affects up to 8 million people worldwide. Parasites are transmitted to human and animal hosts through the bite of an infected sand fly. Novel strategies for disease control require a better understanding of the key step for transmission, namely the establishment of infection inside the fly. Methods The aim of this work was to identify sand fly systemic transcriptomic signatures associated with Leishmania infection. We used next generation sequencing to describe the transcriptome of whole Phlebotomus papatasi sand flies when fed with blood alone (control) or with blood containing one of three trypanosomatids: Leishmania major, L. donovani and Herpetomonas muscarum, the latter being a parasite not transmitted to humans. Results Of the trypanosomatids studied, only L. major was able to successfully establish an infection in the host P. papatasi. However, the transcriptional signatures observed after each parasite-contaminated blood meal were not specific to success or failure of a specific infection and they did not differ from each other. The transcriptional signatures were also indistinguishable after a non-contaminated blood meal. Conclusions The results imply that sand flies perceive Leishmania as just one feature of their microbiome landscape and that any strategy to tackle transmission should focus on the response towards the blood meal rather than parasite establishment. Alternatively, Leishmania could suppress host responses. These results will generate new thinking around the concept of stopping transmission by controlling the parasite inside the insect.![]()
Collapse
Affiliation(s)
- Megan A Sloan
- Department of Biochemistry, University of Oxford, South Parks Rd, Oxford, OX1 3QU, UK
| | - Jovana Sadlova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Tereza Lestinova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Mandy J Sanders
- The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, Cambridgeshire, UK
| | - James A Cotton
- The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, Cambridgeshire, UK
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Petros Ligoxygakis
- Department of Biochemistry, University of Oxford, South Parks Rd, Oxford, OX1 3QU, UK.
| |
Collapse
|
50
|
RÊGO FELIPED, SOARES RODRIGOPEDRO. Lutzomyia longipalpis: an update on this sand fly vector. AN ACAD BRAS CIENC 2021. [DOI: 10.1590/0001-3765202120200254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|