1
|
de Freitas Milagres T, López-de-Felipe M, da Silva WJ, Martín-Martín I, Gálvez R, da Silva OS. Same parasite, different outcomes: unraveling the epidemiology of Leishmania infantum infection in Brazil and Spain. Trends Parasitol 2023; 39:774-785. [PMID: 37442747 DOI: 10.1016/j.pt.2023.06.008] [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: 03/17/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023]
Abstract
Human leishmaniosis caused by Leishmania infantum is an important health problem worldwide. One of the main aspects arousing interest is the epidemiological scenario surrounding Le. infantum infection in the New World (NW) and Old World (OW). This parasite was introduced to the Americas during European colonization leading to different epidemiology outcomes, even more enigmatic in the face of global changes. Thus, this review aims to highlight the differences and similarities between Le. infantum epidemiology between Brazil (NW) and Spain (OW), as both countries are leading the total number of leishmaniosis cases in their respective continents. Grounded on a systemic view, this article also draws attention to possible common innovative strategies to rethink ways of controlling infections caused by Le. infantum.
Collapse
Affiliation(s)
- Tarcísio de Freitas Milagres
- Flebocollect Medical Entomology Citizen Science Group, Madrid, Spain; Department of Microbiology, Immunology and Parasitology, Universidade Federal Do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Wellington Junior da Silva
- Department of Microbiology, Immunology and Parasitology, Universidade Federal Do Rio Grande do Sul, Porto Alegre, Brazil
| | - Inés Martín-Martín
- Laboratory of Medical Entomology, National Center for Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Gálvez
- Flebocollect Medical Entomology Citizen Science Group, Madrid, Spain; Department of Specific Didactics, School of Education and Teacher Training, Universidad Autónoma de Madrid, Madrid, Spain.
| | - Onilda Santos da Silva
- Department of Microbiology, Immunology and Parasitology, Universidade Federal Do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
2
|
Valenzuela-Leon PC, Campos Chagas A, Martin-Martin I, Williams AE, Berger M, Shrivastava G, Paige AS, Kotsyfakis M, Tirloni L, Calvo E. Guianensin, a Simulium guianense salivary protein, has broad anti-hemostatic and anti-inflammatory properties. Front Immunol 2023; 14:1163367. [PMID: 37469515 PMCID: PMC10353047 DOI: 10.3389/fimmu.2023.1163367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/15/2023] [Indexed: 07/21/2023] Open
Abstract
Background Salivary glands from blood-feeding arthropods secrete several molecules that inhibit mammalian hemostasis and facilitate blood feeding and pathogen transmission. The salivary functions from Simulium guianense, the main vector of Onchocerciasis in South America, remain largely understudied. Here, we have characterized a salivary protease inhibitor (Guianensin) from the blackfly Simulium guianense. Materials and methods A combination of bioinformatic and biophysical analyses, recombinant protein production, in vitro and in vivo experiments were utilized to characterize the molecula mechanism of action of Guianensin. Kinetics of Guianensin interaction with proteases involved in vertebrate inflammation and coagulation were carried out by surface plasmon resonance and isothermal titration calorimetry. Plasma recalcification and coagulometry and tail bleeding assays were performed to understand the role of Guianensin in coagulation. Results Guianensin was identified in the sialotranscriptome of adult S. guianense flies and belongs to the Kunitz domain of protease inhibitors. It targets various serine proteases involved in hemostasis and inflammation. Binding to these enzymes is highly specific to the catalytic site and is not detectable for their zymogens, the catalytic site-blocked human coagulation factor Xa (FXa), or thrombin. Accordingly, Guianensin significantly increased both PT (Prothrombin time) and aPTT (Activated partial thromboplastin time) in human plasma and consequently increased blood clotting time ex vivo. Guianensin also inhibited prothrombinase activity on endothelial cells. We show that Guianensin acts as a potent anti-inflammatory molecule on FXa-induced paw edema formation in mice. Conclusion The information generated by this work highlights the biological functionality of Guianensin as an antithrombotic and anti-inflammatory protein that may play significant roles in blood feeding and pathogen transmission.
Collapse
Affiliation(s)
| | - Andrezza Campos Chagas
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, United States
| | - Ines Martin-Martin
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, United States
| | - Adeline E. Williams
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, United States
| | - Markus Berger
- Tick-Pathogen Transmission Unit, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Gaurav Shrivastava
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, United States
| | - Andrew S. Paige
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, United States
| | - Michalis Kotsyfakis
- Laboratory of Genomics and Proteomics of Disease Vectors, Institute of Parasitology, Biology Centre, Czech Academy of Sciences, Ceske Budejovice, Czechia
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Lucas Tirloni
- Tick-Pathogen Transmission Unit, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, Hamilton, MT, United States
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
3
|
Martin-Martin I, Williams AE, Calvo E. Evaluation of Aedes aegypti Feeding Status. Cold Spring Harb Protoc 2023; 2023:pdb.prot108023. [PMID: 36690389 PMCID: PMC10750256 DOI: 10.1101/pdb.prot108023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Female mosquitoes need vertebrate blood for egg development. Evaluating mosquito behavior is essential for determining the ability of a mosquito to blood feed. Blood feeding experiments are often performed using artificial membrane feeders; however, such experiments do not represent realistic scenarios in which a mosquito injects saliva into the host to prevent host hemostatic responses. Vertebrate animal models are therefore more representative of a natural blood feeding event. Here, we describe a methodology to evaluate mosquito blood feeding success that can be used to compare blood feeding between mosquito groups-for instance, wild-type versus transgenic mosquitoes lacking salivary proteins or field-collected versus laboratory-reared mosquitoes. We also include a simple procedure to measure blood meal size, allowing for a more quantitative assessment of feeding status. The volume of ingested blood directly affects mosquito fecundity and fertility, important markers of fitness. The methods described herein can be used to evaluate transmission-blocking vaccines, insecticides, or fitness costs associated with transgenic mosquitoes.
Collapse
Affiliation(s)
- Ines Martin-Martin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852, USA
| | - Adeline E Williams
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852, USA
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852, USA
| |
Collapse
|
4
|
Martin-Martin I, Williams AE, Calvo E. Determination of Mosquito Probing and Feeding Time to Evaluate Mosquito Blood Feeding. Cold Spring Harb Protoc 2023; 2023:pdb.top107659. [PMID: 36669861 PMCID: PMC10751641 DOI: 10.1101/pdb.top107659] [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] [Indexed: 01/22/2023]
Abstract
Hematophagous arthropods are animals that feed on vertebrate blood for egg production. Mosquitoes must pierce the host skin, locate blood vessels, and extract blood without being noticed. Mosquito stylets lacerate host tissues, triggering the activation of the three branches of hemostasis, or stopping of blood flow: vasoconstriction, platelet aggregation, and coagulation. Mosquitoes inject saliva into the host skin during their intradermal search for blood (also called probing), and salivary proteins counteract hemostasis. Blood feeding dynamics have been traditionally described by observational studies, in which researchers using magnifying glasses watched mosquitoes in the act of blood feeding. These studies provided the foundation for protocols to evaluate mosquito blood feeding in a more quantitative manner. Here, we introduce mosquito blood feeding biology with a focus on the feeding steps, which include penetration, probing, and feeding. Understanding blood feeding dynamics is crucial for evaluating probing time and other relevant parameters derived from blood feeding, such as blood meal size, fecundity, and fertility. Other considerations, including the relationship between probing and pathogen transmission and novel technologies to address blood feeding, are also discussed.
Collapse
Affiliation(s)
- Ines Martin-Martin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852, USA
| | - Adeline E Williams
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852, USA
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852, USA
| |
Collapse
|
5
|
Smith LB, Chagas AC, Martin-Martin I, Ribeiro JMC, Calvo E. An insight into the female and male Sabethes cyaneus mosquito salivary glands transcriptome. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 153:103898. [PMID: 36587808 PMCID: PMC9899327 DOI: 10.1016/j.ibmb.2022.103898] [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: 11/29/2022] [Revised: 12/26/2022] [Accepted: 12/26/2022] [Indexed: 06/17/2023]
Abstract
Mosquitoes are responsible for the death and debilitation of millions of people every year due to the pathogens they can transmit while blood feeding. While a handful of mosquitoes, namely those in the Aedes, Anopheles, and Culex genus, are the dominant vectors, many other species belonging to different genus are also involved in various pathogen cycles. Sabethes cyaneus is one of the many poorly understood mosquito species involved in the sylvatic cycle of Yellow Fever Virus. Here, we report the expression profile differences between male and female of Sa.cyaneus salivary glands (SGs). We find that female Sa.cyaneus SGs have 165 up-regulated and 18 down-regulated genes compared to male SGs. Most of the up-regulated genes have unknown functions, however, odorant binding proteins, such as those in the D7 protein family, and mucins were among the top 30 genes. We also performed various in vitro activity assays of female SGs. In the activity analysis we found that female SG extracts inhibit coagulation by blocking factor Xa and has endonuclease activity. Knowledge about mosquitoes and their physiology are important for understanding how different species differ in their ability to feed on and transmits pathogens to humans. These results provide us with an insight into the Sabethes SG activity and gene expression that expands our understanding of mosquito salivary glands.
Collapse
Affiliation(s)
- Leticia Barion Smith
- Laboratory of Malaria and Vector Research, National Institutes of Health, 12735 Twinbrook Parkway, Room 2W09, Bethesda, MD, 20892, USA
| | - Andrezza Campos Chagas
- Laboratory of Malaria and Vector Research, National Institutes of Health, 12735 Twinbrook Parkway, Room 2W09, Bethesda, MD, 20892, USA
| | - Ines Martin-Martin
- Laboratory of Malaria and Vector Research, National Institutes of Health, 12735 Twinbrook Parkway, Room 2W09, Bethesda, MD, 20892, USA
| | - Jose M C Ribeiro
- Laboratory of Malaria and Vector Research, National Institutes of Health, 12735 Twinbrook Parkway, Room 2W09, Bethesda, MD, 20892, USA
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institutes of Health, 12735 Twinbrook Parkway, Room 2W09, Bethesda, MD, 20892, USA.
| |
Collapse
|
6
|
Fayaz S, Bahrami F, Parvizi P, Fard-Esfahani P, Ajdary S. An overview of the sand fly salivary proteins in vaccine development against leishmaniases. IRANIAN JOURNAL OF MICROBIOLOGY 2022; 14:792-801. [PMID: 36721440 PMCID: PMC9867623 DOI: 10.18502/ijm.v14i6.11253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Leishmaniases are a group of vector-borne parasitic diseases transmitted through the infected sand flies. Leishmania parasites are inoculated into the host skin along with sand fly saliva. The sand fly saliva consists of biologically active molecules with anticoagulant, anti-inflammatory, and immunomodulatory properties. Such properties help the parasite circumvent the host's immune responses. The salivary compounds support the survival and multiplication of the parasite and facilitate the disease progression. It is documented that frequent exposure to uninfected sand fly bites produces neutralizing antibodies against specific salivary proteins and further activates the cellular mechanisms to prevent the establishment of the disease. The immune responses due to sand fly saliva are highly specific and depend on the composition of the salivary molecules. Hence, thorough knowledge of these compounds in different sand fly species and information about their antigenicity are paramount to designing an effective vaccine. Herein, we review the composition of the sand fly saliva, immunomodulatory properties of some of its components, immune responses to its proteins, and potential vaccine candidates against leishmaniases.
Collapse
Affiliation(s)
- Shima Fayaz
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Fariborz Bahrami
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Parviz Parvizi
- Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Soheila Ajdary
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,Corresponding author: Soheila Ajdary, Ph.D, Department of Immunology, Pasteur Institute of Iran, Tehran, Iran. Tel: +98-2166968857 Fax: +98-2166968857 ;
| |
Collapse
|
7
|
Kawahori S, Seki C, Mizushima D, Tabbabi A, Yamamoto DS, Kato H. Ayaconin, a novel inhibitor of the plasma contact system from the sand fly Lutzomyia ayacuchensis, a vector of Andean-type cutaneous leishmaniasis. Acta Trop 2022; 234:106602. [PMID: 35817195 DOI: 10.1016/j.actatropica.2022.106602] [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: 06/06/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 11/24/2022]
Abstract
Transcriptome analysis of the salivary gland cDNA library from a phlebotomine sand fly, Lutzomyia ayacuchensis, identified a transcript coding for the PpSP15/SL1 family protein as the second most abundant salivary component. In the present study, a recombinant protein of the PpSP15/SL1 family protein, designated ayaconin, was expressed in Escherichia coli, and its biological activity was characterized. The recombinant ayaconin purified from the soluble fraction of E. coli lysate efficiently inhibited the intrinsic but not extrinsic blood coagulation pathway. When the target of ayaconin was evaluated using fluorescent substrates of coagulation factors, ayaconin inhibited factor XIIa (FXIIa) activity more efficiently in a dose-dependent manner, suggesting that FXII is the primary target of ayaconin. In addition, incubation of ayaconin with FXII prior to activation effectively inhibited FXIIa activity, whereas such inhibition was not observed when ayaconin was mixed after the production of FXIIa, indicating that ayaconin inhibits the activation process of FXII to produce FXIIa, but not the enzymatic activity of FXIIa. Moreover, ayaconin was shown to bind to FXII, suggesting that the binding of ayaconin to FXII is involved in the inhibitory mechanism against FXII activation. These results suggest that ayaconin plays an important role in the blood-sucking of Lu. ayacuchensis.
Collapse
Affiliation(s)
- Satoru Kawahori
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke City, Tochigi 329-0498, Japan
| | - Chisato Seki
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke City, Tochigi 329-0498, Japan
| | - Daiki Mizushima
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke City, Tochigi 329-0498, Japan
| | - Ahmed Tabbabi
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke City, Tochigi 329-0498, Japan
| | - Daisuke S Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke City, Tochigi 329-0498, Japan
| | - Hirotomo Kato
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke City, Tochigi 329-0498, Japan.
| |
Collapse
|
8
|
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: 53] [Impact Index Per Article: 26.5] [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
|
9
|
Aoki V, Abdeladhim M, Li N, Cecilio P, Prisayanh P, Diaz LA, Valenzuela JG. Some Good and Some Bad: Sand Fly Salivary Proteins in the Control of Leishmaniasis and in Autoimmunity. Front Cell Infect Microbiol 2022; 12:839932. [PMID: 35281450 PMCID: PMC8913536 DOI: 10.3389/fcimb.2022.839932] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/02/2022] [Indexed: 01/22/2023] Open
Abstract
Sand flies are hematophagous insects responsible for the transmission of vector-borne diseases to humans. Prominent among these diseases is Leishmaniasis that affects the skin and mucous surfaces and organs such as liver and spleen. Importantly, the function of blood-sucking arthropods goes beyond merely transporting pathogens. The saliva of vectors of disease contains pharmacologically active components that facilitate blood feeding and often pathogen establishment. Transcriptomic and proteomic studies have enumerated the repertoire of sand fly salivary proteins and their potential use for the control of Leishmaniasis, either as biomarkers of vector exposure or as anti-Leishmania vaccines. However, a group of specific sand fly salivary proteins triggers formation of cross-reactive antibodies that bind the ectodomain of human desmoglein 1, a member of the epidermal desmosomal cadherins. These cross-reactive antibodies are associated with skin autoimmune blistering diseases, such as pemphigus, in certain immunogenetically predisposed individuals. In this review, we focus on two different aspects of sand fly salivary proteins in the context of human disease: The good, which refers to salivary proteins functioning as biomarkers of exposure or as anti-Leishmania vaccines, and the bad, which refers to salivary proteins as environmental triggers of autoimmune skin diseases.
Collapse
Affiliation(s)
- Valeria Aoki
- Department of Dermatology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Universidade de Sao Paulo, Sao Paulo, Brazil
- *Correspondence: Valeria Aoki,
| | - Maha Abdeladhim
- 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
| | - Ning Li
- Department of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Pedro Cecilio
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Phillip Prisayanh
- Department of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Luis A. Diaz
- Department of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - 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
| |
Collapse
|
10
|
Volpedo G, Huston RH, Holcomb EA, Pacheco-Fernandez T, Gannavaram S, Bhattacharya P, Nakhasi HL, Satoskar AR. From infection to vaccination: reviewing the global burden, history of vaccine development, and recurring challenges in global leishmaniasis protection. Expert Rev Vaccines 2021; 20:1431-1446. [PMID: 34511000 DOI: 10.1080/14760584.2021.1969231] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Leishmaniasis is a major public health problem and the second most lethal parasitic disease in the world due to the lack of effective treatments and vaccines. Even when not lethal, leishmaniasis significantly affects individuals and communities through life-long disabilities, psycho-sociological trauma, poverty, and gender disparity in treatment. AREAS COVERED This review discusses the most relevant and recent research available on Pubmed and GoogleScholar highlighting leishmaniasis' global impact, pathogenesis, treatment options, and lack of effective control strategies. An effective vaccine is necessary to prevent morbidity and mortality, lower health care costs, and reduce the economic burden of leishmaniasis for endemic low- and middle-income countries. Since there are several forms of leishmaniasis, a pan-Leishmania vaccine without geographical restrictions is needed. This review also focuses on recent advances and common challenges in developing prophylactic strategies against leishmaniasis. EXPERT OPINION Despite advances in pre-clinical vaccine research, approval of a human leishmaniasis vaccine still faces major challenges - including manufacturing of candidate vaccines under Good Manufacturing Practices, developing well-designed clinical trials suitable in endemic countries, and defined correlates of protection. In addition, there is a need to explore Challenge Human Infection Model to avoid large trials because of fluctuating incidence and prevalence of leishmanasis.
Collapse
Affiliation(s)
- Greta Volpedo
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Ryan H Huston
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Erin A Holcomb
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Thalia Pacheco-Fernandez
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Parna Bhattacharya
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Hira L Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Abhay R Satoskar
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
11
|
Schneider CA, Calvo E, Peterson KE. Arboviruses: How Saliva Impacts the Journey from Vector to Host. Int J Mol Sci 2021; 22:ijms22179173. [PMID: 34502092 PMCID: PMC8431069 DOI: 10.3390/ijms22179173] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/19/2021] [Accepted: 08/22/2021] [Indexed: 12/21/2022] Open
Abstract
Arthropod-borne viruses, referred to collectively as arboviruses, infect millions of people worldwide each year and have the potential to cause severe disease. They are predominately transmitted to humans through blood-feeding behavior of three main groups of biting arthropods: ticks, mosquitoes, and sandflies. The pathogens harbored by these blood-feeding arthropods (BFA) are transferred to animal hosts through deposition of virus-rich saliva into the skin. Sometimes these infections become systemic and can lead to neuro-invasion and life-threatening viral encephalitis. Factors intrinsic to the arboviral vectors can greatly influence the pathogenicity and virulence of infections, with mounting evidence that BFA saliva and salivary proteins can shift the trajectory of viral infection in the host. This review provides an overview of arbovirus infection and ways in which vectors influence viral pathogenesis. In particular, we focus on how saliva and salivary gland extracts from the three dominant arbovirus vectors impact the trajectory of the cellular immune response to arbovirus infection in the skin.
Collapse
Affiliation(s)
- Christine A. Schneider
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA;
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA;
| | - Karin E. Peterson
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA;
- Correspondence:
| |
Collapse
|
12
|
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: 43] [Impact Index Per Article: 14.3] [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
|
13
|
Karmakar S, Nath S, Sarkar B, Chakraborty S, Paul S, Karan M, Pal C. Insect vectors' saliva and gut microbiota as a blessing in disguise: probability versus possibility. Future Microbiol 2021; 16:657-670. [PMID: 34100305 DOI: 10.2217/fmb-2020-0239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Drawing of host blood is a natural phenomenon during the bite of blood-probing insect vectors. Along with the blood meal, the vectors introduce salivary components and a trail of microbiota. In the case of infected vectors, the related pathogen accompanies the aforementioned biological components. In addition to Anopheles gambiae or Anopheles stephensi, the bites of other nonmalarial vectors cannot be ignored in malaria-endemic regions. Similarly, the bite incidence of Phlebotomus papatasi cannot be ignored in visceral leishmaniasis-endemic regions. Even the chances of getting bitten by uninfected vectors are higher than the infected vectors. We have discussed the probability or possibility of uninfected, infected, and/or nonvector's saliva and gut microbiota as a therapeutic option leading to the initial deterrent to pathogen establishment.
Collapse
Affiliation(s)
- Suman Karmakar
- Cellular Immunology & Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India.,Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India
| | - Supriya Nath
- Cellular Immunology & Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India.,Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India
| | - Biswajyoti Sarkar
- Cellular Immunology & Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India.,Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India
| | - Sondipon Chakraborty
- Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India
| | - Sharmistha Paul
- Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India
| | - Mintu Karan
- Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India
| | - Chiranjib Pal
- Cellular Immunology & Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India.,Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India
| |
Collapse
|
14
|
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: 2.3] [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
|
15
|
Kern O, Valenzuela Leon PC, Gittis AG, Bonilla B, Cruz P, Chagas AC, Ganesan S, Ribeiro JMC, Garboczi DN, Martin-Martin I, Calvo E. The structures of two salivary proteins from the West Nile vector Culex quinquefasciatus reveal a beta-trefoil fold with putative sugar binding properties. Curr Res Struct Biol 2021; 3:95-105. [PMID: 34235489 PMCID: PMC8244437 DOI: 10.1016/j.crstbi.2021.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 01/22/2023] Open
Abstract
Female mosquitoes require blood meals for egg development. The saliva of blood feeding arthropods contains biochemically active molecules, whose anti-hemostatic and anti-inflammatory properties facilitate blood feeding on vertebrate hosts. While transcriptomics has presented new opportunities to investigate the diversity of salivary proteins from hematophagous arthropods, many of these proteins remain functionally undescribed. Previous transcriptomic analysis of female salivary glands from Culex quinquefasciatus, an important vector of parasitic and viral infections, uncovered a 12-member family of putatively secreted proteins of unknown function, named the Cysteine and Tryptophan-Rich (CWRC) proteins. Here, we present advances in the characterization of two C. quinquefasciatus CWRC family members, CqDVP-2 and CqDVP-4, including their enrichment in female salivary glands, their specific localization within salivary gland tissues, evidence that these proteins are secreted into the saliva, and their native crystal structures, at 2.3 Å and 1.87 Å, respectively. The β-trefoil fold common to CqDVP-2 and CqDVP-4 is similar to carbohydrate-binding proteins, including the B subunit of the AB toxin, ricin, from the castor bean Ricinus communis. Further, we used a glycan array approach, which identifies carbohydrate ligands associated with inflammatory processes and signal transduction. Glycan array 300 testing identified 100 carbohydrate moieties with positive binding to CqDVP-2, and 77 glycans with positive binding to CqDVP-4. The glycan with the highest relative fluorescence intensities, which exhibited binding to both CqDVP-2 and CqDVP-4, was used for molecular docking experiments. We hypothesize that these proteins bind to carbohydrates on the surface of cells important to host immunology. Given that saliva is deposited into the skin during a mosquito bite, and acts as the vehicle for arbovirus inoculation, understanding the role of these proteins in pathogen transmission is of critical importance. This work presents the first solved crystal structures of C. quinquefasciatus salivary proteins with unknown function. These two molecules are the second and third structures reported from salivary proteins from C. quinquefasciatus, an important, yet understudied disease vector.
Collapse
Affiliation(s)
- Olivia Kern
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Paola Carolina Valenzuela Leon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Apostolos G Gittis
- Structural Biology Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Brian Bonilla
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Phillip Cruz
- Bioinformatics and Computational Biosciences Branch. Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Andrezza Campos Chagas
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Sundar Ganesan
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Jose M C Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - David N Garboczi
- Structural Biology Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Ines Martin-Martin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| |
Collapse
|
16
|
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
|
17
|
Sugitani K, Egorova D, Mizumoto S, Nishio S, Yamada S, Kitagawa H, Oshima K, Nadano D, Matsuda T, Miyata S. Hyaluronan degradation and release of a hyaluronan-aggrecan complex from perineuronal nets in the aged mouse brain. Biochim Biophys Acta Gen Subj 2020; 1865:129804. [PMID: 33253804 DOI: 10.1016/j.bbagen.2020.129804] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/16/2020] [Accepted: 11/24/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Perineuronal nets (PNNs) are insoluble aggregates of extracellular matrix molecules in the brain that consist of hyaluronan (HA) and chondroitin sulfate proteoglycans (CSPGs). PNNs promote the acquisition and storage of memories by stabilizing the formation of synapses in the adult brain. Although the deterioration of PNNs has been suggested to contribute to the age-dependent decline in brain function, the molecular mechanisms underlying age-related changes in PNNs remain unclear. METHODS The amount and solubility of PNN components were investigated by sequential extraction followed by a disaccharide analysis and immunoblotting. We examined the interaction between HA and aggrecan, a major HA-binding CSPG, by combining mass spectrometry and pull-down assays. RESULTS The solubility and amount of HA increased in the brain with age. Among several CSPGs, the solubility of aggrecan was selectively elevated during aging. In contrast to alternations in biochemical properties, the expression of PNN components at the transcript level was not markedly changed by aging. The increased solubility of aggrecan was not due to the loss of HA-binding properties. Our results indicated that the degradation of high-molecular-mass HA induced the release of the HA-aggrecan complex from PNNs in the aged brain. CONCLUSION The present study revealed a novel mechanism underlying the age-related deterioration of PNNs in the brain.
Collapse
Affiliation(s)
- Kei Sugitani
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Diana Egorova
- Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwaicho, Fuchu, Tokyo 183-8509, Japan
| | - Shuji Mizumoto
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-Ku, Nagoya 468-8503, Japan
| | - Shunsuke Nishio
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Shuhei Yamada
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-Ku, Nagoya 468-8503, Japan
| | - Hiroshi Kitagawa
- Laboratory of Biochemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada-Ku, Kobe 658-8558, Japan
| | - Kenzi Oshima
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Daita Nadano
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Tsukasa Matsuda
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan; Faculty of Food and Agricultural Sciences, Fukushima University, Kanayagawa 1, Fukushima 960-1296, Japan
| | - Shinji Miyata
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan; Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwaicho, Fuchu, Tokyo 183-8509, Japan.
| |
Collapse
|
18
|
Martin-Martin I, Kern O, Brooks S, Smith LB, Valenzuela-Leon PC, Bonilla B, Ackerman H, Calvo E. Biochemical characterization of AeD7L2 and its physiological relevance in blood feeding in the dengue mosquito vector, Aedes aegypti. FEBS J 2020; 288:2014-2029. [PMID: 32799410 DOI: 10.1111/febs.15524] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/23/2020] [Accepted: 08/12/2020] [Indexed: 01/02/2023]
Abstract
Aedes aegypti saliva facilitates blood meal acquisition through pharmacologically active compounds that prevent host hemostasis. Among these salivary proteins are the D7s, which are highly abundant and have been shown to act as scavengers of biogenic amines and eicosanoids. In this work, we performed comparative structural modeling, characterized the binding capabilities, and assessed the physiological functions of the Ae. aegypti salivary protein AeD7L2 compared to the well-characterized AeD7L1. AeD7L1 and AeD7L2 show different binding affinities to several biogenic amines and biolipids involved in host hemostasis. Interestingly, AeD7L2 tightly binds U-46619, the stable analog of thromboxane A2 (KD = 69.4 nm), which is an important platelet aggregation mediator, while AeD7L1 shows no binding. We tested the ability of these proteins to interfere with the three branches of hemostasis: vasoconstriction, platelet aggregation, and blood coagulation. Pressure myography experiments showed these two proteins reversed isolated resistance artery vasoconstriction induced by either norepinephrine or U-46619. These proteins also inhibited platelet aggregation induced by low doses of collagen or U-46619. However, D7 long proteins did not affect blood coagulation. The different ligand specificity and affinities of AeD7L1 and AeD7L2 matched our experimental observations from studying their effects on vasoconstriction and platelet aggregation, which confirm their role in preventing host hemostasis. This work highlights the complex yet highly specific biological activities of mosquito salivary proteins and serves as another example of the sophisticated biology underlying arthropod blood feeding.
Collapse
Affiliation(s)
- Ines Martin-Martin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Olivia Kern
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Steven Brooks
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Leticia Barion Smith
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Paola Carolina Valenzuela-Leon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Brian Bonilla
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Hans Ackerman
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
19
|
Tirado TC, de Andrade AJ, Ribeiro MCVDC, Figueiredo FB. Use of the high-content imaging system equipment to evaluate in vitro infection by Leishmania braziliensis in response to sand fly Nyssomyia neivai saliva. Acta Trop 2020; 209:105540. [PMID: 32442434 DOI: 10.1016/j.actatropica.2020.105540] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/13/2020] [Accepted: 05/13/2020] [Indexed: 12/13/2022]
Abstract
Earlier research has shown that in vivo immunization with sand fly saliva protects the host against infection by parasites of genus Leishmania, and inoculation of saliva along with Leishmania promastigotes favors infection in the host. In this study, High-Content Imaging System was used to demonstrate in vitro that sand fly saliva also promotes infection by these parasites. THP-1 cells were cultured in 96-well microplates and challenged with three strains of Leishmania braziliensis plus four dilutions of Nyssomyia neivai salivary gland extract. High-Content Imaging System equipment (Operetta CLS, Perkin Elmer) was configured to automatically count both cells and parasites inside the microplates and subsequently calculate the Infection Index (II). Results demonstrate that the extract concentration of 1 gland showed greater infection than other dilutions. These findings suggest that sand fly N. neivai saliva has potential for increasing the parasite infection, reinforcing the importance of studying its components. A new method to evaluate Leishmania infection in vitro assays was also presented, broadening this area of study.
Collapse
Affiliation(s)
- Thais Cristina Tirado
- Laboratório de Parasitologia Molecular, Departamento de Patologia Básica, Universidade Federal do Paraná (UFPR), Curitiba, Paraná, Brazil; Laboratório de Biologia Celular, Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ), Curitiba, Paraná, Brazil.
| | - Andrey José de Andrade
- Laboratório de Parasitologia Molecular, Departamento de Patologia Básica, Universidade Federal do Paraná (UFPR), Curitiba, Paraná, Brazil
| | | | - Fabiano Borges Figueiredo
- Laboratório de Biologia Celular, Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ), Curitiba, Paraná, Brazil
| |
Collapse
|
20
|
Takechi M, Oshima K, Nadano D, Kitagawa H, Matsuda T, Miyata S. A pericellular hyaluronan matrix is required for the morphological maturation of cortical neurons. Biochim Biophys Acta Gen Subj 2020; 1864:129679. [PMID: 32623025 DOI: 10.1016/j.bbagen.2020.129679] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Hyaluronan (HA) is a major component of the extracellular matrix (ECM) and is involved in many cellular functions. In the adult brain, HA forms macromolecular aggregates around synapses and plays important roles in neural plasticity. In contrast to the well-characterized function of HA in the adult brain, its roles in the developing brain remain largely unknown. METHODS Biochemical and histochemical analyses were performed to analyze the amount, solubility, and localization of HA in the developing mouse brain. By combining in utero labeling, cell isolation, and in vitro cultures, we examined the expression of hyaluronan synthase (HAS) and morphological maturation of cortical neurons. RESULTS The amount of HA increased during perinatal development and decreased in the adult. HA existed as a soluble form in the early stages; however, its solubility markedly decreased during postnatal development. HA localized in cell-sparse regions in the embryonic stages, but was broadly distributed during the postnatal development of the cerebral cortex. Developing cortical neurons expressed both Has2 and Has3, but not Has1, suggesting the autonomous production of HA by neurons themselves. HA formed a pericellular matrix around the cell bodies and neurites of developing cortical neurons, and the inhibition of HA synthesis reduced neurite outgrowth. CONCLUSION The formation of the pericellular HA matrix is essential for the proper morphological maturation of developing neurons. GENERAL SIGNIFICANCE This study provides new insights into the roles of hyaluronan in the brain. DEVELOPMENT
Collapse
Affiliation(s)
- Mina Takechi
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Kenzi Oshima
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Daita Nadano
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Hiroshi Kitagawa
- Laboratory of Biochemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada-Ku, Kobe 658-8558, Japan
| | - Tsukasa Matsuda
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan; Faculty of Food and Agricultural Sciences, Fukushima University, Kanayagawa 1, Fukushima 960-1296, Japan
| | - Shinji Miyata
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan; Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwaicho, Fuchu, Tokyo 183-8509, Japan.
| |
Collapse
|
21
|
Martin-Martin I, Paige A, Valenzuela Leon PC, Gittis AG, Kern O, Bonilla B, Chagas AC, Ganesan S, Smith LB, Garboczi DN, Calvo E. ADP binding by the Culex quinquefasciatus mosquito D7 salivary protein enhances blood feeding on mammals. Nat Commun 2020; 11:2911. [PMID: 32518308 PMCID: PMC7283271 DOI: 10.1038/s41467-020-16665-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 05/13/2020] [Indexed: 12/26/2022] Open
Abstract
During blood-feeding, mosquito saliva is injected into the skin to facilitate blood meal acquisition. D7 proteins are among the most abundant components of the mosquito saliva. Here we report the ligand binding specificity and physiological relevance of two D7 long proteins from Culex quinquefasciatus mosquito, the vector of filaria parasites or West Nile viruses. CxD7L2 binds biogenic amines and eicosanoids. CxD7L1 exhibits high affinity for ADP and ATP, a binding capacity not reported in any D7. We solve the crystal structure of CxD7L1 in complex with ADP to 1.97 Å resolution. The binding pocket lies between the two protein domains, whereas all known D7s bind ligands either within the N- or the C-terminal domains. We demonstrate that these proteins inhibit hemostasis in ex vivo and in vivo experiments. Our results suggest that the ADP-binding function acquired by CxD7L1 evolved to enhance blood-feeding in mammals, where ADP plays a key role in platelet aggregation. D7 proteins are highly abundant in the salivary glands of several blood feeding insects. Here, the authors study the ligand binding specificity and physiological roles of the mosquito D7 proteins CxD7L1 and CxD7L2, showing that CxD7L1 acquired ADP-binding properties to enhance blood feeding in mammals.
Collapse
Affiliation(s)
- Ines Martin-Martin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Andrew Paige
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Paola Carolina Valenzuela Leon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Apostolos G Gittis
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Olivia Kern
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Brian Bonilla
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Andrezza Campos Chagas
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Sundar Ganesan
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Leticia Barion Smith
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - David N Garboczi
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA.
| |
Collapse
|
22
|
Polanska N, Ishemgulova A, Volfova V, Flegontov P, Votypka J, Yurchenko V, Volf P. Sergentomyia schwetzi: Salivary gland transcriptome, proteome and enzymatic activities in two lineages adapted to different blood sources. PLoS One 2020; 15:e0230537. [PMID: 32208452 PMCID: PMC7092997 DOI: 10.1371/journal.pone.0230537] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 03/02/2020] [Indexed: 11/18/2022] Open
Abstract
During the blood feeding, sand fly females inject saliva containing immunomodulatory and anti-haemostatic molecules into their vertebrate hosts. The saliva composition is species-specific, likely due to an adaptation to particular haemostatic pathways of their preferred host. Research on sand fly saliva is limited to the representatives of two best-studied genera, Phlebotomus and Lutzomyia. Although the members of the genus Sergentomyia are highly abundant in many areas in the Old World, their role in human disease transmission remains uncertain. Most Sergentomyia spp. preferentially attack various species of reptiles, but feeding on warm-blooded vertebrates, including humans and domestic animals, has been repeatedly described, especially for Sergentomyia schwetzi, of which salivary gland transcriptome and proteome is analyzed in the current study. Illumina RNA sequencing and de novo assembly of the reads and their annotation revealed 17,293 sequences homologous to other arthropods’ proteins. In the sialome, all proteins typical for sand fly saliva were identified–antigen 5-related, lufaxin, yellow-related, PpSP15-like, D7-related, ParSP25-like, and silk proteins, as well as less frequent salivary proteins included 71kDa-like, ParSP80-like, SP16-like, and ParSP17-like proteins. Salivary enzymes include apyrase, hyaluronidase, endonuclease, amylase, lipase A2, adenosine deaminase, pyrophosphatase, 5’nucleotidase, and ribonuclease. Proteomics analysis of salivary glands identified 631 proteins, 81 of which are likely secreted into the saliva. We also compared two S. schwetzi lineages derived from the same origin. These lineages were adapted for over 40 generations for blood feeding either on mice (S-M) or geckos (S-G), two vertebrate hosts with different haemostatic mechanisms. Altogether, 20 and 40 annotated salivary transcripts were up-regulated in the S-M and S-G lineage, respectively. Proteomic comparison revealed ten salivary proteins more abundant in the lineage S-M, whereas 66 salivary proteins were enriched in the lineage S-G. No difference between lineages was found for apyrase activity; contrarily the hyaluronidase activity was significantly higher in the lineage feeding on mice.
Collapse
Affiliation(s)
- Nikola Polanska
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
- * E-mail:
| | - Aygul Ishemgulova
- Life Science Research Centre, Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Vera Volfova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Pavel Flegontov
- Life Science Research Centre, Faculty of Science, University of Ostrava, Ostrava, Czech Republic
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, České Budějovice (Budweis), Czech Republic
| | - Jan Votypka
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Vyacheslav Yurchenko
- Life Science Research Centre, Faculty of Science, University of Ostrava, Ostrava, Czech Republic
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, České Budějovice (Budweis), Czech Republic
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector Borne Diseases, Sechenov University, Moscow, Russia
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
23
|
Coutinho De Oliveira B, Duthie MS, Alves Pereira VR. Vaccines for leishmaniasis and the implications of their development for American tegumentary leishmaniasis. Hum Vaccin Immunother 2019; 16:919-930. [PMID: 31634036 DOI: 10.1080/21645515.2019.1678998] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The leishmaniases are a collection of vector-borne parasitic diseases caused by a number of different Leishmania species that are distributed worldwide. Clinical and laboratory research have together revealed several important immune components that control Leishmania infection and indicate the potential of immunization to prevent leishmaniasis. In this review we introduce previous and ongoing experimental research efforts to develop vaccines against Leishmania species. First, second and third generation vaccine strategies that have been proposed to counter cutaneous and visceral leishmaniasis (CL and VL, respectively) are summarized. One of the major bottlenecks in development is the transition from results in animal model studies to humans, and we highlight that although American tegumentary leishmaniasis (ATL; New World CL) can progress to destructive and disfiguring mucosal lesions, most research has been conducted using mouse models and Old World Leishmania species. We conclude that assessment of vaccine candidates in ATL settings therefore appears merited.
Collapse
Affiliation(s)
- Beatriz Coutinho De Oliveira
- Pós-Graduação em Inovação Terapêutica, Universidade Federal de Pernambuco (UFPE), Recife, Brazil.,Departamento de Imunologia, Instituto Aggeu Magalhães, Recife, Brazil
| | | | | |
Collapse
|
24
|
Yang Z, Wu Y. Improved annotation of Lutzomyia longipalpis genome using bioinformatics analysis. PeerJ 2019; 7:e7862. [PMID: 31616601 PMCID: PMC6790103 DOI: 10.7717/peerj.7862] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 09/10/2019] [Indexed: 01/09/2023] Open
Abstract
Lutzomyia longipalpis, a sand fly, is a vector-spreading pathogenic protozoan in the New World. MicroRNA (miRNA) is evolutionarily-conserved non-coding RNA, which plays critical roles in various biological processes. To date, the functions of most proteins in L. longipalpis are unknown, and few studies have addressed the roles of miRNAs in this species. In the present study, we re-annotated the protein-coding genes and identified several miRNAs using a set of comparative genomics tools. A large number of L. longipalpis proteins were found to be homologous with those in the mosquito genome, indicating that they may have experienced similar selective pressures. Among these proteins, a set of 19 putative salivary proteins were identified, which could be used for studying the transmission of Leishmania. Twenty-one novel miRNAs were characterized, including two miRNAs, miR-4113-5p and miR-5101, which are unique to L. longipalpis. Many of the targets of these two genes were found to be involved in ATP hydrolysis-coupled proton transport, suggesting that they may have important roles in the physiology of energy production. Topology analysis of the miRNA-gene network indicated that miR-9388-5p and miR-3871-5p regulate several critical genes in response to disease development. In conclusion, our work provides a basis for improving the genome annotation of L. longipalpis, and opens a new door to understanding the molecular regulatory mechanisms in this species.
Collapse
Affiliation(s)
- Zhiyuan Yang
- College of Life Information Science & Instrument Engineering, Hangzhou Dianzi University, Hangzhou, PR China
| | - Ying Wu
- College of Chemical Engineering, Huaqiao University, Xiamen, PR China
| |
Collapse
|
25
|
Need for sustainable approaches in antileishmanial drug discovery. Parasitol Res 2019; 118:2743-2752. [DOI: 10.1007/s00436-019-06443-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 08/23/2019] [Indexed: 12/16/2022]
|
26
|
Volfova V, Volf P. The salivary hyaluronidase and apyrase of the sand fly Sergentomyia schwetzi (Diptera, Psychodidae). INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2018; 102:67-74. [PMID: 30273670 DOI: 10.1016/j.ibmb.2018.09.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/26/2018] [Accepted: 09/27/2018] [Indexed: 06/08/2023]
Abstract
Current knowledge of sand fly salivary components has been based solely on Lutzomyia and Phlebotomus species which feed mainly on mammals; their hyaluronidases and apyrases were demonstrated to significantly affect blood meal intake and transmission of vector-borne pathogens. Members of the third sand fly genus Sergentomyia preferentially feed on reptiles but some of them are considered as Leishmania and arboviruses vectors; however, nothing is known about their salivary components that might be relevant for pathogens transmission. Here, marked hyaluronidase and apyrase activities were demonstrated in the saliva of a Sergentomyia schwetzi colony maintained on geckos. Hyaluronidase of S. schwetzi cleaved hyaluronan as the prominent substrate, and was active over a broad pH range from 4.0 to 8.0, with a sharp peak at pH 5.0. SDS PAGE zymography demonstrated the monomeric character of the enzyme, which remained active in reducing conditions. The apparent molecular weight of 43 kDa was substantially lower than in any sand fly species tested so far and may indicate relatively low grade of the glycosylation of the enzyme. The apyrase of S. schwetzi was typical strictly Ca2+ dependent Cimex-family apyrase. It was active over a pH range from 6.5 to 9.0, with a peak of activity at pH 8.5, and had an ATPase/ADPase ratio of 0.9. The apyrase activity increased during the first 3 days post-emergence, then reached a plateau and remained relatively constant until day 8. In comparison with a majority of Phlebotomus and Lutzomyia species tested to date, both the hyaluronidase and apyrase activities of S. schwetzi were relatively low, which may reflect an adaptation of this sand fly to blood feeding on non-mammalian hosts.
Collapse
Affiliation(s)
- Vera Volfova
- Department of Parasitology, Faculty of Science, Charles University, Vinicna 7, Prague 2, 128 44, Czech Republic
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Vinicna 7, Prague 2, 128 44, Czech Republic.
| |
Collapse
|