1
|
Wang Y, Ling L, Jiang L, Marin-Lopez A. Research progress toward arthropod salivary protein vaccine development for vector-borne infectious diseases. PLoS Negl Trop Dis 2024; 18:e0012618. [PMID: 39636798 PMCID: PMC11620354 DOI: 10.1371/journal.pntd.0012618] [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: 12/07/2024] Open
Abstract
Hematophagous arthropods, including mosquitoes, ticks, and flies, are responsible for the transmission of several pathogens to vertebrates 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. In general, attempts to develop vaccines for pathogens transmitted by arthropods have met with moderate success, with few vaccine candidates currently developed. Nowadays, there are vaccine candidates under clinical trials, including different platforms, like mRNA, DNA, recombinant viral vector-based, virus-like particles (VLPs), inactivated-virus, live-attenuated virus, peptide and protein-based vaccines, all of them based on the presentation of pathogen antigens to the host immune system. A new approach to prevent VBDs has arose during the last decades, based on the design of vaccines that target vector-derived antigens. The salivary secretions of arthropods, in addition of causing allergic reactions and harbor pathogens, are also involved in the transmission and infection establishment in the host, altering its immune responses. In this review, we summarize the achievements in the arthropod salivary-based vaccine development for different vector-borne infectious diseases. This provides a rationale for creating vaccines against different types of arthropod salivary proteins, such as mosquitoes, ticks, and sand flies. Using salivary proteins of clinically important vectors might contribute to achieve protection against and control multiple arthropod-borne infection diseases.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Inspection and Quarantine, Shanghai Customs College, Shanghai, China
| | - Lin Ling
- Department of Inspection and Quarantine, Shanghai Customs College, Shanghai, China
| | - Lijie Jiang
- Department of Inspection and Quarantine, Shanghai Customs College, Shanghai, China
| | - Alejandro Marin-Lopez
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
2
|
Sghaier RM, Benhnini F, Guerfali FZ, Attia H, Bali A, Zaatour A, Mkannez G, Gharbi A, Belhaj-Hamida N, Dridi H, Ben-Salah A, Dellagi K, Laouini D. Healed Lesions of Human Cutaneous Leishmaniasis Caused By Leishmania major Do Not Shelter Persistent Residual Parasites. Front Cell Infect Microbiol 2022; 12:839216. [PMID: 35967864 PMCID: PMC9363604 DOI: 10.3389/fcimb.2022.839216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 04/15/2022] [Indexed: 11/13/2022] Open
Abstract
In human cutaneous leishmaniasis (HCL) caused by Leishmania (L.) major, the cutaneous lesions heal spontaneously and induce a Th1-type immunity that confers solid protection against reinfection. The same holds true for the experimental leishmaniasis induced by L. major in C57BL/6 mice where residual parasites persist after spontaneous clinical cure and induce sustainable memory immune responses and resistance to reinfection. Whether residual parasites also persist in scars of cured HCL caused by L. major is still unknown. Cutaneous scars from 53 volunteers with healed HCL caused by L. major were biopsied and the tissue sample homogenates were analyzed for residual parasites by four methods: i) microscope detection of amastigotes, ii) parasite culture by inoculation on biphasic medium, iii) inoculation of tissue exctracts to the footpad of BALB/c mice, an inbred strain highly susceptible to L. major, and iv) amplification of parasite kDNA by a highly sensitive real-time PCR (RT-PCR). Our results show that the scars of healed lesions of HCL caused by L. major do not contain detectable residual parasites, suggesting that this form likely induces a sterile cure at least within the scars. This feature contrasts with other Leishmania species causing chronic, diffuse, or recidivating forms of leishmaniasis where parasites do persist in healed lesions. The possibility that alternative mechanisms to parasite persistence are needed to boost and maintain long-term immunity to L. major, should be taken into consideration in vaccine development against L. major infection.
Collapse
Affiliation(s)
- Rabiaa M. Sghaier
- LR16IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
| | - Fouad Benhnini
- LR16IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
| | - Fatma Z. Guerfali
- LR16IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
| | - Hanène Attia
- LR16IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
| | - Aymen Bali
- LR16IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
| | - Amor Zaatour
- LR16IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
- Service of Medical Epidemiology, Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
| | - Ghada Mkannez
- LR16IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
| | - Adel Gharbi
- LR16IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
- Service of Medical Epidemiology, Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
| | - Nabil Belhaj-Hamida
- LR16IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
- Service of Medical Epidemiology, Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
| | - Hichem Dridi
- LR16IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
- Service of Medical Epidemiology, Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
| | - Afif Ben-Salah
- LR16IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
- Service of Medical Epidemiology, Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Department of Family and Community Medicine, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Koussay Dellagi
- LR16IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
| | - Dhafer Laouini
- LR16IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Institut Pasteur de Tunis, Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
- *Correspondence: Dhafer Laouini, ;
| |
Collapse
|
3
|
Machado AS, Lage DP, Vale DL, Freitas CS, Linhares FP, Cardoso JM, Pereira IA, Ramos FF, Tavares GS, Ludolf F, Oliveira-da-Silva JA, Bandeira RS, Simões AC, Duarte MC, Oliveira JS, Christodoulides M, Chávez-Fumagalli MA, Roatt BM, Martins VT, Coelho EA. A recombinant Leishmania amastigote-specific protein, rLiHyG, with adjuvants, protects against infection with Leishmania infantum. Acta Trop 2022; 230:106412. [PMID: 35305943 DOI: 10.1016/j.actatropica.2022.106412] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/11/2022]
Abstract
Vaccination against visceral leishmaniasis (VL) should be considered as a control measure to protect against disease, and amastigote-specific proteins could help to develop such vaccines, since this parasite form is in contact with the host immune system during the active disease. In this study, a Leishmania amastigote-specific protein, LiHyG, was evaluated as recombinant protein (rLiHyG) as vaccine candidate against Leishmania infantum infection in BALB/c mice. The protein was associated with saponin (rLiHyG/Sap) or Poloxamer 407-based polymeric micelles (rLiHyG/Mic) as adjuvants, and animals receiving saline, saponin or micelle as controls. Immunological and parasitological analyses were performed before (n = 8 per group; as primary endpoint) and after (n = 8 per group; as secondary endpoint) infection. Results showed that, in both endpoints, rLiHyG/Sap and rLiHyG/Mic induced higher levels of IFN-γ, IL-12 and GM-CSF in spleen cell cultures from vaccinated animals, besides elevated presence of IgG2a isotype antibodies. Decreased hepatotoxicity and 'positive lymphoproliferative response were also found after challenge. Such findings reflected in significantly lower levels of parasite load found in their spleens, livers, bone marrows and draining lymph nodes. In conclusion, rLiHyG associated with Th1-type adjuvant could be considered for future studies as vaccine candidate to protect against VL.
Collapse
|
4
|
Haghdoust S, Noroozbeygi M, Hajimollahoseini M, Masooleh MM, Yeganeh F. A candidate vaccine composed of live nonpathogenic Iranian Lizard Leishmania mixed with Chitin microparticles protects mice against Leishmania major infection. Acta Trop 2022; 227:106298. [PMID: 34971566 DOI: 10.1016/j.actatropica.2021.106298] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 12/26/2021] [Accepted: 12/27/2021] [Indexed: 11/01/2022]
Abstract
BACKGROUND The protective effect of immunization using Iranian Lizard Leishmania (ILL) mixed with CpG oligodeoxynucleotides (CpG-ODN) was demonstrated in a previous study. Here, we report the effect of leishmanization using ILL mixed with chitin microparticles (CMPs) as an adjuvant against L. major infection in BALB/c mice. METHODS Briefly, 2 × 107 live ILL were mixed with 10 µg CMPs (<40 μm in size) (ILL+CMP) and were injected subcutaneously into the right footpad of BALB/c mice. Three control groups were included in the study and received ILL, chitin, and PBS respectively. Three weeks later, mice were challenged with 2 × 105 live L. majorEGFP promastigotes, which were inoculated into the left footpad. The infection course was monitored using footpad swelling measurement and in vivo imaging. Eleven weeks after the challenge, all mice were sacrificed and parasite burden was measured in the spleen and the draining lymph node using three different methods including real-time PCR, flow cytometry, and direct fluorescent microscopy. In addition, cytokines levels (IFN-γ and IL-10), and nitric oxide production were assayed in splenocytes. RESULTS Mice immunized with ILL+CMP had a smaller footpad diameter in comparison to control groups and notably, no lesion was developed at the inoculation site. Additionally, in vivo imaging study revealed that there was no detectable fluorescence in the ILL+CMP group footpad by the end of the tenth week. This finding was confirmed by three methods used for parasite burden assays. Moreover, higher IFN-γ level was observed in mice immunized with ILL+CMP in comparison with other groups. On the other hand, nitric oxide concentration was higher in the ILL control group. CONCLUSION ILL mixed with chitin microparticles is an effective vaccine against leishmaniasis in BALB/c mice. This vaccine is able to induce an adequate immune response to decrease the parasite burden and prevent lesion formation. Further studies are needed to evaluate long-lasting immunity, especially in experimental outbreed models.
Collapse
|
5
|
Chang KP, Reynolds JM, Liu Y, He JJ. Leishmaniac Quest for Developing a Novel Vaccine Platform. Is a Roadmap for Its Advances Provided by the Mad Dash to Produce Vaccines for COVID-19? Vaccines (Basel) 2022; 10:vaccines10020248. [PMID: 35214706 PMCID: PMC8874365 DOI: 10.3390/vaccines10020248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 02/04/2023] Open
Abstract
“Bugs as drugs” in medicine encompasses the use of microbes to enhance the efficacy of vaccination, such as the delivery of vaccines by Leishmania—the protozoan etiological agent of leishmaniasis. This novel approach is appraised in light of the successful development of vaccines for Covid-19. All relevant aspects of this pandemic are summarized to provide the necessary framework in contrast to leishmaniasis. The presentation is in a side-by-side matching format with particular emphasis on vaccines. The comparative approach makes it possible to highlight the timeframe of the vaccine workflows condensed by the caveats of pandemic urgency and, at the same time, provides the background of Leishmania behind its use as a vaccine carrier. Previous studies in support of the latter are summarized as follows. Leishmaniasis confers life-long immunity on patients after cure, suggesting the effective vaccination is achievable with whole-cell Leishmania. A new strategy was developed to inactivate these cells in vitro, rendering them non-viable, hence non-disease causing, albeit retaining their immunogenicity and adjuvanticity. This was achieved by installing a dual suicidal mechanism in Leishmania for singlet oxygen (1O2)-initiated inactivation. In vitro cultured Leishmania were genetically engineered for cytosolic accumulation of UV-sensitive uroporphyrin I and further loaded endosomally with a red light-sensitive cationic phthalocyanine. Exposing these doubly dye-loaded Leishmania to light triggers intracellular production of highly reactive but extremely short-lived 1O2, resulting in their rapid and complete inactivation. Immunization of susceptible animals with such inactivated Leishmania elicited immunity to protect them against experimental leishmaniasis. Significantly, the inactivated Leishmania was shown to effectively deliver transgenically add-on ovalbumin (OVA) to antigen-presenting cells (APC), wherein OVA epitopes were processed appropriately for presentation with MHC molecules to activate epitope-specific CD8+ T cells. Application of this approach to deliver cancer vaccine candidates, e.g., enolase-1, was shown to suppress tumor development in mouse models. A similar approach is predicted to elicit lasting immunity against infectious diseases, including complementation of the spike protein-based vaccines in use for COVID-19. This pandemic is devastating, but brings to light the necessity of considering many facets of the disease in developing vaccination programs. Closer collaboration is essential among those in diverse disciplinary areas to provide the roadmap toward greater success in the future. Highlighted herein are several specific issues of vaccinology and new approaches worthy of consideration due to the pandemic.
Collapse
|
6
|
Kaye PM, Mohan S, Mantel C, Malhame M, Revill P, Le Rutte E, Parkash V, Layton AM, Lacey CJ, Malvolti S. Overcoming roadblocks in the development of vaccines for leishmaniasis. Expert Rev Vaccines 2021; 20:1419-1430. [PMID: 34727814 PMCID: PMC9844205 DOI: 10.1080/14760584.2021.1990043] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/04/2021] [Indexed: 01/21/2023]
Abstract
INTRODUCTION The leishmaniases represent a group of parasitic diseases caused by infection with one of several species of Leishmania parasites. Disease presentation varies because of differences in parasite and host genetics and may be influenced by additional factors such as host nutritional status or co-infection. Studies in experimental models of Leishmania infection, vaccination of companion animals and human epidemiological data suggest that many forms of leishmaniasis could be prevented by vaccination, but no vaccines are currently available for human use. AREAS COVERED We describe some of the existing roadblocks to the development and implementation of an effective leishmaniasis vaccine, based on a review of recent literature found on PubMed, BioRxiv and MedRxiv. In addition to discussing scientific unknowns that hinder vaccine candidate identification and selection, we explore gaps in knowledge regarding the commercial and public health value propositions underpinning vaccine development and provide a route map for future research and advocacy. EXPERT OPINION Despite significant progress, leishmaniasis vaccine development remains hindered by significant gaps in understanding that span the vaccine development pipeline. Increased coordination and adoption of a more holistic view to vaccine development will be required to ensure more rapid progress in the years ahead.
Collapse
Affiliation(s)
- Paul M. Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, Heslington, York, UK
| | - Sakshi Mohan
- Centre for Health Economics, University of York, Heslington, York, UK
| | | | | | - Paul Revill
- Centre for Health Economics, University of York, Heslington, York, UK
| | - Epke Le Rutte
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Vivak Parkash
- York Biomedical Research Institute, Hull York Medical School, University of York, Heslington, York, UK
| | - Alison M. Layton
- York Biomedical Research Institute, Hull York Medical School, University of York, Heslington, York, UK
| | - Charles J.N. Lacey
- York Biomedical Research Institute, Hull York Medical School, University of York, Heslington, York, UK
| | | |
Collapse
|
7
|
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: 27] [Impact Index Per Article: 6.8] [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
|
8
|
Demarta-Gatsi C, Mécheri S. Vector saliva controlled inflammatory response of the host may represent the Achilles heel during pathogen transmission. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20200155. [PMID: 34035796 PMCID: PMC8128132 DOI: 10.1590/1678-9199-jvatitd-2020-0155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Infection with vector-borne pathogens starts with the inoculation of these pathogens during blood feeding. In endemic regions, the population is regularly bitten by naive vectors, implicating a permanent stimulation of the immune system by the vector saliva itself (pre-immune context). Comparatively, the number of bites received by exposed individuals from non-infected vectors is much higher than the bites from infected ones. Therefore, vector saliva and the immunological response in the skin may play an important role, so far underestimated, in the establishment of anti-pathogen immunity in endemic areas. Hence, the parasite biology and the disease pathogenesis in “saliva-primed” and “saliva-unprimed” individuals must be different. This integrated view on how the pathogen evolves within the host together with vector salivary components, which are known to be endowed with a variety of pharmacological and immunological properties, must remain the focus of any investigational study dealing with vector-borne diseases. Considering this three-way partnership, the host skin (immune system), the pathogen, and the vector saliva, the approach that consists in the validation of vector saliva as a source of molecular entities with anti-disease vaccine potential has been recently a subject of active and fruitful investigation. As an example, the vaccination with maxadilan, a potent vasodilator peptide extracted from the saliva of the sand fly Lutzomyia longipalpis, was able to protect against infection with various leishmanial parasites. More interestingly, a universal mosquito saliva vaccine that may potentially protect against a range of mosquito-borne infections including malaria, dengue, Zika, chikungunya and yellow fever. In this review, we highlight the key role played by the immunobiology of vector saliva in shaping the outcome of vector-borne diseases and discuss the value of studying diseases in the light of intimate cross talk among the pathogen, the vector saliva, and the host immune mechanisms.
Collapse
Affiliation(s)
- Claudia Demarta-Gatsi
- Institut Pasteur, Unité de Biologie des Interactions Hôte Parasites, Paris, France.,CNRS ERL9195, Paris, France.,INSERM U1201, Paris, France.,Medicines for Malaria Venture (MMV), Geneva, Switzerland.,Institut Pasteur, Unité de Biologie des Interactions Hôte Parasites, Paris, France
| | - Salah Mécheri
- Institut Pasteur, Unité de Biologie des Interactions Hôte Parasites, Paris, France.,CNRS ERL9195, Paris, France.,INSERM U1201, Paris, France
| |
Collapse
|
9
|
Younis BM, Osman M, Khalil EAG, Santoro F, Furini S, Wiggins R, Keding A, Carraro M, Musa AEA, Abdarahaman MAA, Mandefield L, Bland M, Aebischer T, Gabe R, Layton AM, Lacey CJN, Kaye PM, Musa AM. Safety and immunogenicity of ChAd63-KH vaccine in post-kala-azar dermal leishmaniasis patients in Sudan. Mol Ther 2021; 29:2366-2377. [PMID: 33781913 PMCID: PMC8261165 DOI: 10.1016/j.ymthe.2021.03.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/06/2020] [Accepted: 03/23/2021] [Indexed: 11/25/2022] Open
Abstract
Post-kala-azar dermal leishmaniasis (PKDL) is a chronic, stigmatizing skin condition occurring frequently after apparent clinical cure from visceral leishmaniasis. Given an urgent need for new treatments, we conducted a phase IIa safety and immunogenicity trial of ChAd63-KH vaccine in Sudanese patients with persistent PKDL. LEISH2a (ClinicalTrials.gov: NCT02894008) was an open-label three-phase clinical trial involving sixteen adult and eight adolescent patients with persistent PKDL (median duration, 30 months; range, 6-180 months). Patients received a single intramuscular vaccination of 1 × 1010 viral particles (v.p.; adults only) or 7.5 × 1010 v.p. (adults and adolescents), with primary (safety) and secondary (clinical response and immunogenicity) endpoints evaluated over 42-120 days follow-up. AmBisome was provided to patients with significant remaining disease at their last visit. ChAd63-KH vaccine showed minimal adverse reactions in PKDL patients and induced potent innate and cell-mediated immune responses measured by whole-blood transcriptomics and ELISpot. 7/23 patients (30.4%) monitored to study completion showed >90% clinical improvement, and 5/23 (21.7%) showed partial improvement. A logistic regression model applied to blood transcriptomic data identified immune modules predictive of patients with >90% clinical improvement. A randomized controlled trial to determine whether these clinical responses were vaccine-related and whether ChAd63-KH vaccine has clinical utility is underway.
Collapse
Affiliation(s)
- Brima M Younis
- Department of Clinical Pathology & Immunology, Institute of Endemic Diseases, University of Khartoum, Army Ave., Khartoum, Sudan
| | - Mohamed Osman
- York Biomedical Research Institute, Hull York Medical School, University of York, Heslington, York YO10 5DD, UK
| | - Eltahir A G Khalil
- Department of Clinical Pathology & Immunology, Institute of Endemic Diseases, University of Khartoum, Army Ave., Khartoum, Sudan
| | - Francesco Santoro
- Department of Medical Biotechnologies, University of Siena, Siena 53100, Italy
| | - Simone Furini
- Department of Medical Biotechnologies, University of Siena, Siena 53100, Italy
| | - Rebecca Wiggins
- York Biomedical Research Institute, Hull York Medical School, University of York, Heslington, York YO10 5DD, UK
| | - Ada Keding
- Department of Health Sciences, University of York, Heslington, York YO10 5DD, UK
| | - Monica Carraro
- Department of Medical Biotechnologies, University of Siena, Siena 53100, Italy
| | - Anas E A Musa
- Department of Clinical Pathology & Immunology, Institute of Endemic Diseases, University of Khartoum, Army Ave., Khartoum, Sudan
| | - Mujahid A A Abdarahaman
- Department of Clinical Pathology & Immunology, Institute of Endemic Diseases, University of Khartoum, Army Ave., Khartoum, Sudan
| | - Laura Mandefield
- Department of Health Sciences, University of York, Heslington, York YO10 5DD, UK
| | - Martin Bland
- Department of Health Sciences, University of York, Heslington, York YO10 5DD, UK
| | | | - Rhian Gabe
- Wolfson Institute of Preventive Medicine, Queen Mary University of London, London E1 4NS, UK
| | - Alison M Layton
- York Biomedical Research Institute, Hull York Medical School, University of York, Heslington, York YO10 5DD, UK
| | - Charles J N Lacey
- York Biomedical Research Institute, Hull York Medical School, University of York, Heslington, York YO10 5DD, UK
| | - Paul M Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, Heslington, York YO10 5DD, UK.
| | - Ahmed M Musa
- Department of Clinical Pathology & Immunology, Institute of Endemic Diseases, University of Khartoum, Army Ave., Khartoum, Sudan.
| |
Collapse
|
10
|
Abstract
Efforts to produce vaccines against SARS and MERS were prematurely halted since their scope was perceived to be geographically restricted and they were subsequently categorized as neglected diseases. However, when a similar virus spread globally triggering the COVID-19 pandemic, we were harshly reminded that several other neglected diseases might also be waiting for the perfect opportunity to become mainstream. As climate change drives urbanization, natural selection of pathogens and their intermediate vectors and reservoirs, the risk of neglected diseases emerging within a larger susceptible pool becomes an even greater threat. Availability of a vaccine for COVID-19 is widely considered the only way to end this pandemic. Similarly, vaccines are also seen as the best tools available to control the spread of neglected (sometimes referred to as emerging or re-emerging) diseases, until the water, hygiene and sanitation infrastructure is improved in areas of their prevalence. Vaccine production is usually cost and labour intensive and thus minimal funding is directed towards controlling and eliminating neglected diseases (NDs). A customised but sustainable approach is needed to develop and deploy vaccines against NDs. While safety, efficacy and public trust are the three main success pillars for most vaccines, affordability is vital when formulating vaccines for neglected diseases.
Collapse
|
11
|
Pinart M, Rueda JR, Romero GA, Pinzón-Flórez CE, Osorio-Arango K, Silveira Maia-Elkhoury AN, Reveiz L, Elias VM, Tweed JA. Interventions for American cutaneous and mucocutaneous leishmaniasis. Cochrane Database Syst Rev 2020; 8:CD004834. [PMID: 32853410 PMCID: PMC8094931 DOI: 10.1002/14651858.cd004834.pub3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND On the American continent, cutaneous and mucocutaneous leishmaniasis (CL and MCL) are diseases associated with infection by several species of Leishmania parasites. Pentavalent antimonials remain the first-choice treatment. There are alternative interventions, but reviewing their effectiveness and safety is important as availability is limited. This is an update of a Cochrane Review first published in 2009. OBJECTIVES To assess the effects of interventions for all immuno-competent people who have American cutaneous and mucocutaneous leishmaniasis (ACML). SEARCH METHODS We updated our database searches of the Cochrane Skin Group Specialised Register, CENTRAL, MEDLINE, Embase, LILACS and CINAHL to August 2019. We searched five trials registers. SELECTION CRITERIA Randomised controlled trials (RCTs) assessing either single or combination treatments for ACML in immuno-competent people, diagnosed by clinical presentation and Leishmania infection confirmed by smear, culture, histology, or polymerase chain reaction on a biopsy specimen. The comparators were either no treatment, placebo only, or another active compound. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. Our key outcomes were the percentage of participants 'cured' at least three months after the end of treatment, adverse effects, and recurrence. We used GRADE to assess evidence certainty for each outcome. MAIN RESULTS We included 75 studies (37 were new), totalling 6533 randomised participants with ATL. The studies were mainly conducted in Central and South America at regional hospitals, local healthcare clinics, and research centres. More male participants were included (mean age: roughly 28.9 years (SD: 7.0)). The most common confirmed species were L. braziliensis, L. panamensis, and L. mexicana. The most assessed interventions and comparators were non-antimonial systemics (particularly oral miltefosine) and antimonials (particularly meglumine antimoniate (MA), which was also a common intervention), respectively. Three studies included moderate-to-severe cases of mucosal leishmaniasis but none included cases with diffuse cutaneous or disseminated CL, considered the severe cutaneous form. Lesions were mainly ulcerative and located in the extremities and limbs. The follow-up (FU) period ranged from 28 days to 7 years. All studies had high or unclear risk of bias in at least one domain (especially performance bias). None of the studies reported the degree of functional or aesthetic impairment, scarring, or quality of life. Compared to placebo, at one-year FU, intramuscular (IM) MA given for 20 days to treat L. braziliensis and L. panamensis infections in ACML may increase the likelihood of complete cure (risk ratio (RR) 4.23, 95% confidence interval (CI) 0.84 to 21.38; 2 RCTs, 157 participants; moderate-certainty evidence), but may also make little to no difference, since the 95% CI includes the possibility of both increased and reduced healing (cure rates), and IMMA probably increases severe adverse effects such as myalgias and arthralgias (RR 1.51, 95% CI 1.17 to 1.96; 1 RCT, 134 participants; moderate-certainty evidence). IMMA may make little to no difference to the recurrence risk, but the 95% CI includes the possibility of both increased and reduced risk (RR 1.79, 95% CI 0.17 to 19.26; 1 RCT, 127 participants; low-certainty evidence). Compared to placebo, at six-month FU, oral miltefosine given for 28 days to treat L. mexicana, L. panamensis and L. braziliensis infections in American cutaneous leishmaniasis (ACL) probably improves the likelihood of complete cure (RR 2.25, 95% CI 1.42 to 3.38), and probably increases nausea rates (RR 3.96, 95% CI 1.49 to 10.48) and vomiting (RR 6.92, 95% CI 2.68 to 17.86) (moderate-certainty evidence). Oral miltefosine may make little to no difference to the recurrence risk (RR 2.97, 95% CI 0.37 to 23.89; low-certainty evidence), but the 95% CI includes the possibility of both increased and reduced risk (all based on 1 RCT, 133 participants). Compared to IMMA, at 6 to 12 months FU, oral miltefosine given for 28 days to treat L. braziliensis, L. panamensis, L. guyanensis and L. amazonensis infections in ACML may make little to no difference to the likelihood of complete cure (RR 1.05, 95% CI 0.90 to 1.23; 7 RCTs, 676 participants; low-certainty evidence). Based on moderate-certainty evidence (3 RCTs, 464 participants), miltefosine probably increases nausea rates (RR 2.45, 95% CI 1.72 to 3.49) and vomiting (RR 4.76, 95% CI 1.82 to 12.46) compared to IMMA. Recurrence risk was not reported. For the rest of the key comparisons, recurrence risk was not reported, and risk of adverse events could not be estimated. Compared to IMMA, at 6 to 12 months FU, oral azithromycin given for 20 to 28 days to treat L. braziliensis infections in ACML probably reduces the likelihood of complete cure (RR 0.51, 95% CI 0.34 to 0.76; 2 RCTs, 93 participants; moderate-certainty evidence). Compared to intravenous MA (IVMA) and placebo, at 12 month FU, adding topical imiquimod to IVMA, given for 20 days to treat L. braziliensis, L. guyanensis and L. peruviana infections in ACL probably makes little to no difference to the likelihood of complete cure (RR 1.30, 95% CI 0.95 to 1.80; 1 RCT, 80 participants; moderate-certainty evidence). Compared to MA, at 6 months FU, one session of local thermotherapy to treat L. panamensis and L. braziliensis infections in ACL reduces the likelihood of complete cure (RR 0.80, 95% CI 0.68 to 0.95; 1 RCT, 292 participants; high-certainty evidence). Compared to IMMA and placebo, at 26 weeks FU, adding oral pentoxifylline to IMMA to treat CL (species not stated) probably makes little to no difference to the likelihood of complete cure (RR 0.86, 95% CI 0.63 to 1.18; 1 RCT, 70 participants; moderate-certainty evidence). AUTHORS' CONCLUSIONS Evidence certainty was mostly moderate or low, due to methodological shortcomings, which precluded conclusive results. Overall, both IMMA and oral miltefosine probably result in an increase in cure rates, and nausea and vomiting are probably more common with miltefosine than with IMMA. Future trials should investigate interventions for mucosal leishmaniasis and evaluate recurrence rates of cutaneous leishmaniasis and its progression to mucosal disease.
Collapse
Affiliation(s)
- Mariona Pinart
- Free time independent Cochrane reviewer, Berlin, Germany
| | - José-Ramón Rueda
- Department of Preventive Medicine and Public Health, University of the Basque Country, Leioa, Spain
| | - Gustavo As Romero
- Center for Tropical Medicine, University of Brasilia, Brasilia, Brazil
| | | | - Karime Osorio-Arango
- Dirección de Redes en Salud Pública, Instituto Nacional de Salud, Bogotá, Colombia
| | - Ana Nilce Silveira Maia-Elkhoury
- Communicable Diseases and Environmental Determinants of Health (CDE), Neglected, Tropical and Vector Borne Diseases (VT), Pan American Health Organization/ World Health Organization (PAHO/WHO), Rio de Janeiro, Brazil
| | - Ludovic Reveiz
- Evidence and Intelligence for Action in Health Department, Pan American Health Organization (PAHO), Washington DC, USA
| | - Vanessa M Elias
- Evidence and Intelligence for Action in Health Department, Pan American Health Organization (PAHO), Washington DC, USA
| | - John A Tweed
- c/o Cochrane Skin Group, The University of Nottingham, Nottingham, UK
| |
Collapse
|
12
|
Oliveira-da-Silva JA, Lage DP, Ramos FF, Machado AS, Tavares GS, Mendonça DV, Pereira IA, Martins VT, Carvalho LM, Ludolf F, Santos TT, Reis TA, Oliveira CS, Bandeira RS, Silva AM, Costa LE, Oliveira JS, Duarte MC, Menezes-Souza D, Roatt BM, Teixeira AL, Coelho EA. Leishmania infantum pyridoxal kinase evaluated in a recombinant protein and DNA vaccine to protects against visceral leishmaniasis. Mol Immunol 2020; 124:161-171. [DOI: 10.1016/j.molimm.2020.06.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/04/2020] [Accepted: 06/11/2020] [Indexed: 01/10/2023]
|
13
|
Affiliation(s)
- Pat Nuttall
- Department of Zoology, University of Oxford, Oxford OX1 3SZ, UK.
| |
Collapse
|
14
|
Chimeric Vaccines Designed by Immunoinformatics-Activated Polyfunctional and Memory T Cells That Trigger Protection against Experimental Visceral Leishmaniasis. Vaccines (Basel) 2020; 8:vaccines8020252. [PMID: 32471081 PMCID: PMC7349981 DOI: 10.3390/vaccines8020252] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/20/2022] Open
Abstract
Many vaccine candidates against visceral leishmaniasis (VL) have been proposed; however, to date, none of them have been efficacious for the human or canine disease. On this basis, the design of leishmaniasis vaccines has been constantly changing, and the use of approaches to select specific epitopes seems to be crucial in this scenario. The ability to predict T cell-specific epitopes makes immunoinformatics an even more necessary approach, as in VL an efficient immune response against the parasite is triggered by T lymphocytes in response to Leishmania spp. immunogenic antigens. Moreover, the success of vaccines depends on the capacity to generate long-lasting memory and polyfunctional cells that are able to eliminate the parasite. In this sense, our study used a combination of different approaches to develop potential chimera candidate vaccines against VL. The first point was to identify the most immunogenic epitopes of Leishmania infantum proteins and construct chimeras composed of Major histocompatibility complex (MHC) class I and II epitopes. For this, we used immunoinformatics features. Following this, we validated these chimeras in a murine model in a thorough memory study and multifunctionality of T cells that contribute to a better elucidation of the immunological protective mechanisms of polyepitope vaccines (chimera A and B) using multicolor flow cytometry. Our results showed that in silico-designed chimeras can elicit polyfunctional T cells producing T helper (Th)1 cytokines, a strong immune response against Leishmania antigen, and the generation of central and effector memory T cells in the spleen cells of vaccinated animals that was able to reduce the parasite burden in this organ. These findings contribute two potential candidate vaccines against VL that can be used in further studies, and help in this complex field of vaccine development against this challenging parasite.
Collapse
|
15
|
Inflammation following trypanosome infection and persistence in the skin. Curr Opin Immunol 2020; 66:65-73. [PMID: 32446136 DOI: 10.1016/j.coi.2020.04.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/10/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023]
Abstract
Human African trypanosomes rely for their transmission on tsetse flies (Glossina sp.) that inoculate parasites into the skin during blood feeding. The absence of a protective vaccine, limited knowledge about the infection immunology, and the existence of asymptomatic carriers sustaining transmission are major outstanding challenges towards elimination. All these relate to the skin where (i) parasites persist and transmit to tsetse flies and (ii) a successful vaccination strategy should ideally be effective. Host immune processes and parasite strategies that underlie early infection and skin tropism are essential aspects to comprehend the transmission-success of trypanosomes and the failure in vaccine development. Recent insights into the early infection establishment may pave the way to novel strategies aimed at blocking transmission.
Collapse
|
16
|
Evaluation of immune response to Bacillus subtilis spores expressing Clonorchis sinensis serpin3. Parasitology 2020; 147:1080-1087. [PMID: 32404215 DOI: 10.1017/s0031182020000797] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Clonorchis sinensis (C. sinensis) is one of the most serious food-borne parasites, which can lead to liver fibrosis or cholangiocarcinoma. Effective measures for clonorchiasis prevention are still urgently needed. Bacillus subtilis (B. subtilis) is an effective antigen delivery platform for oral vaccines. Chonorchis sinensis serpin (CsSerpin) was proved to be potential vaccine candidates. In this study, CsSerpin3 was displayed on the surface of B. subtilis spore and recombinant spores were orally administrated to BALB/C mice. CsSerpin3-specific IgA levels in faecal, bile and intestinal mucous increased at 4-8 weeks after the first administration compared with those in control groups. The mucus production and the number of goblet cells in intestinal mucosa elevated in B.s-CotC-CsSerpin3 (CotC, coat protein of B. subtilis spore) spores treated group compared to those in blank control. No significant difference in the activities of glutamic-pyruvic transaminase/ alanine aminotransferase and glutamic oxalacetic transaminase/aspartate aminotransferase were observed between groups. There was no side effect inflammation and observable pathological damage in the liver tissue of mice after administration. Moreover, collagen deposition and Ishak score were statistically reduced in B.s-CotC-CsSerpin3 spores treated mice. In conclusion, B. subtilis spores displaying CsSerpin3 could be investigated further as an oral vaccine against clonorchiasis.
Collapse
|
17
|
Immune Profile of the Nasal Mucosa in Patients with Cutaneous Leishmaniasis. Infect Immun 2020; 88:IAI.00881-19. [PMID: 32094254 DOI: 10.1128/iai.00881-19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 02/10/2020] [Indexed: 11/20/2022] Open
Abstract
Localized skin lesions are characteristic of cutaneous leishmaniasis (CL); however, Leishmania (Viannia) species, which are responsible for most CL cases in the Americas, can spread systemically, sometimes resulting in mucosal disease. Detection of Leishmania has been documented in healthy mucosal tissues (conjunctiva, tonsils, and nasal mucosa) and healthy skin of CL patients and in individuals with asymptomatic infection in areas of endemicity of L (V) panamensis and L (V) braziliensis transmission. However, the conditions and mechanisms that favor parasite persistence in healthy mucosal tissues are unknown. In this descriptive study, we compared the cell populations of the nasal mucosa (NM) of healthy donors and patients with active CL and explored the immune gene expression signatures related to molecular detection of Leishmania in this tissue in the absence of clinical signs or symptoms of mucosal disease. The cellular composition and gene expression profiles of NM samples from active CL patients were similar to those of healthy volunteers, with a predominance of epithelial over immune cells, and within the CD45+ cell population, a higher frequency of CD66b+ followed by CD14+ and CD3+ cells. In CL patients with molecular evidence of Leishmania persistence in the NM, genes characteristic of an anti-inflammatory and tissue repair responses (IL4R, IL5RA, POSTN, and SATB1) were overexpressed relative to NM samples from CL patients in which Leishmania was not detected. Here, we report the first immunological description of subclinically infected NM tissues of CL patients and provide evidence of a local anti-inflammatory environment favoring parasite persistence in the NM.
Collapse
|
18
|
Mohammed ASA, Tian W, Zhang Y, Peng P, Wang F, Li T. Leishmania lipophosphoglycan components: A potent target for synthetic neoglycoproteins as a vaccine candidate for leishmaniasis. Carbohydr Polym 2020; 237:116120. [PMID: 32241437 DOI: 10.1016/j.carbpol.2020.116120] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 02/21/2020] [Accepted: 03/03/2020] [Indexed: 11/27/2022]
Abstract
Leishmania is an obligate intracellular pathogen that invades phagocytic host cells. Due to its high morbidity and mortality rates, leishmaniasis attracts significant attention. The disease, which is caused by Leishmania parasites, is distributed worldwide, particularly among developing communities, and causes fatal complications if not treated expediently. Unfortunately, the existing treatments are not preventive and do not impede Leishmania infection. Many drugs available for leishmaniasis are becoming less effective due to emerging resistance in some Leishmania species. Other drugs have drawbacks such as low cost-effectiveness, toxicity, and side effects. The World Health Organization (WHO) considers leishmaniasis to be a major public health problem and suggests that the best prevention is to develop a vaccine for this dangerous disease. In this review, we focus on the unique components of lipophosphoglycan (LPG), a component of the Leishmania cell wall, particularly [Galp(1 → 4)-β-[Manp-(1 → 2)-α-Manp-(1 → 2)-α]-Manp] in the cryptic tetrasaccharide cap, and on synthetic approaches as a potent candidate for a leishmaniasis vaccine.
Collapse
Affiliation(s)
- Aiman Saleh A Mohammed
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong, China; National Glycoengineering Research Center, Shandong University, Jinan, 250012, Shandong, China
| | - Weilu Tian
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong, China
| | - Youqin Zhang
- National Glycoengineering Research Center, Shandong University, Jinan, 250012, Shandong, China
| | - Peng Peng
- National Glycoengineering Research Center, Shandong University, Jinan, 250012, Shandong, China
| | - Fengshan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong, China; National Glycoengineering Research Center, Shandong University, Jinan, 250012, Shandong, China.
| | - Tianlu Li
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong, China; National Glycoengineering Research Center, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
19
|
Davarpanah E, Seyed N, Bahrami F, Rafati S, Safaralizadeh R, Taheri T. Lactococcus lactis expressing sand fly PpSP15 salivary protein confers long-term protection against Leishmania major in BALB/c mice. PLoS Negl Trop Dis 2020; 14:e0007939. [PMID: 31899767 PMCID: PMC6941807 DOI: 10.1371/journal.pntd.0007939] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 11/20/2019] [Indexed: 02/06/2023] Open
Abstract
Cutaneous leishmaniasisis a vector-borne disease transmitted by Leishmania infected sand flies. PpSP15 is an immunogenic salivary protein from the sand fly Phlebotomus papatasi. Immunization with PpSP15 was shown to protect against Leishmania major infection. Lactococcus lactis is a safe non-pathogenic delivery system that can be used to express antigens in situ. Here, the codon-optimized Ppsp15-egfp gene was cloned in pNZ8121 vector downstream of the PrtP signal peptide that is responsible for expression and secretion of the protein on the cell wall. Expression of PpSP15-EGFP recombinant protein was monitored by immunofluorescence, flow cytometry and Western blot. Also, expression of protein in cell wall compartment was verified using whole cell ELISA, Western blot and TEM microscopy. BALB/c mice were immunized three times with recombinant L. lactis-PpSP15-EGFPcwa, and the immune responses were followed up, at short-term (ST, 2 weeks) and long-term (LT, 6 months) periods. BALB/c mice were challenged with L. major plus P. papatasi Salivary Gland Homogenate. Evaluation of footpad thickness and parasite burden showed a delay in the development of the disease and significantly decreased parasite numbers in PpSP15 vaccinated animals as compared to control group. In addition, immunized mice showed Th1 type immune responses. Importantly, immunization with L. lactis-PpSP15-EGFPcwa stimulated the long-term memory in mice which lasted for at least 6 months.
Collapse
Affiliation(s)
- Elaheh Davarpanah
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Negar Seyed
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Fariborz Bahrami
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Safaralizadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Tahereh Taheri
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
20
|
A new multi-epitope peptide vaccine induces immune responses and protection against Leishmania infantum in BALB/c mice. Med Microbiol Immunol 2019; 209:69-79. [PMID: 31696313 DOI: 10.1007/s00430-019-00640-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 10/15/2019] [Indexed: 02/06/2023]
Abstract
Visceral leishmaniasis (VL) is a tropical and subtropical disease which is endemic in more than eighty countries around the world. Leishmania infantum is one of the main causative agents of VL disease. Currently, there is no approved-to-market vaccine for VL therapy. In this study, we evaluated cellular and humoral immune responses induced by our newly designed multi-epitope vaccine in BALB/c mice. Four antigenic proteins, including histone H1, sterol 24-c-methyltransferase (SMT), Leishmania-specific hypothetical protein (LiHy), and Leishmania-specific antigenic protein (LSAP) were chosen for the prediction of potential immunodominant epitopes. Moreover, to enhance vaccine immunogenicity, two toll-like receptors 4 (TLR4) agonists, resuscitation-promoting factors of Mycobacterium tuberculosis (RpfE and RpfB), were employed as the built-in adjuvants. Immunization with the designed multi-epitope vaccine elicited a robust Th1-type immune response, compared to other groups, as shown by increased levels of IL-2, IFN-γ, TNF-α, and IgG2a. Furthermore, a significant decrease was observed in Th-2-type-related cytokines such as IL-4 in immunized mice. The designed construct also induced a significant reduction in parasite load (p < 0.0001), conferring protection against L. infantum challenge. This study could be promising in gaining insight towards the potential of peptide epitope-based vaccines as effective protective approaches against Leishmania species.
Collapse
|
21
|
Manning JE, Morens DM, Kamhawi S, Valenzuela JG, Memoli M. Mosquito Saliva: The Hope for a Universal Arbovirus Vaccine? J Infect Dis 2019; 218:7-15. [PMID: 29617849 DOI: 10.1093/infdis/jiy179] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 03/28/2018] [Indexed: 02/06/2023] Open
Abstract
Arthropod-borne viruses (arboviruses) are taxonomically diverse causes of significant morbidity and mortality. In recent decades, important mosquito-borne viruses such as West Nile, chikungunya, dengue, and Zika have re-emerged and spread widely, in some cases pandemically, to cause serious public health emergencies. There are no licensed vaccines against most of these viruses, and vaccine development and use has been complicated by the number of different viruses to protect against, by subtype and strain variation, and by the inability to predict when and where outbreaks will occur. A new approach to preventing arboviral diseases is suggested by the observation that arthropod saliva facilitates transmission of pathogens, including leishmania parasites, Borrelia burgdorferi, and some arboviruses. Viruses carried within mosquito saliva may more easily initiate host infection by taking advantage of the host's innate and adaptive immune responses to saliva. This provides a rationale for creating vaccines against mosquito salivary proteins, rather than against only the virus proteins contained within the saliva. As proof of principle, immunization with sand fly salivary antigens to prevent leishmania infection has shown promising results in animal models. A similar approach using salivary proteins of important vector mosquitoes, such as Aedes aegypti, might protect against multiple mosquito-borne viral infections.
Collapse
Affiliation(s)
- Jessica E Manning
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, Maryland.,Laboratory of Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - David M Morens
- Office of the Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Shaden Kamhawi
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, Maryland
| | - Jesus G Valenzuela
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, Maryland
| | - Matthew Memoli
- Laboratory of Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
22
|
Hohman LS, Peters NC. CD4 + T Cell-Mediated Immunity against the Phagosomal Pathogen Leishmania: Implications for Vaccination. Trends Parasitol 2019; 35:423-435. [PMID: 31080088 DOI: 10.1016/j.pt.2019.04.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 12/31/2022]
Abstract
The generation of an efficacious vaccine that elicits protective CD4+ T cell-mediated immunity has been elusive. The lack of a vaccine against the Leishmania parasite is particularly perplexing as infected individuals acquire life-long immunity to reinfection. Experimental observations suggest that the relationship between immunological memory and protection against Leishmania is not straightforward and that a new paradigm is required to inform vaccine design. These observations include: (i) induction of Th1 memory is a component of protective immunity, but is not sufficient; (ii) memory T cells may be protective only if they generate circulating effector cells prior to, not after, challenge; and (iii) the low-dose/high-inflammation conditions of physiological vector transmission compromises vaccine efficacy. Understanding the implications of these observations is likely key to efficacious vaccination.
Collapse
Affiliation(s)
- Leah S Hohman
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, AB, T2N 4Z6, Canada
| | - Nathan C Peters
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, AB, T2N 4Z6, Canada.
| |
Collapse
|
23
|
Tick saliva and its role in pathogen transmission. Wien Klin Wochenschr 2019; 135:165-176. [PMID: 31062185 PMCID: PMC10118219 DOI: 10.1007/s00508-019-1500-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 04/09/2019] [Indexed: 12/31/2022]
Abstract
Tick saliva is a complex mixture of peptidic and non-peptidic molecules that aid engorgement. The composition of tick saliva changes as feeding progresses and the tick counters the dynamic host response. Ixodid ticks such as Ixodes ricinus, the most important tick species in Europe, transmit numerous pathogens that cause debilitating diseases, e.g. Lyme borreliosis and tick-borne encephalitis. Tick-borne pathogens are transmitted in tick saliva during blood feeding; however, saliva is not simply a medium enabling pathogen transfer. Instead, tick-borne pathogens exploit saliva-induced modulation of host responses to promote their transmission and infection, so-called saliva-assisted transmission (SAT). Characterization of the saliva factors that facilitate SAT is an active area of current research. Besides providing new insights into how tick-borne pathogens survive in nature, the research is opening new avenues for vaccine development.
Collapse
|
24
|
Jablonka W, Kim IH, Alvarenga PH, Valenzuela JG, Ribeiro JMC, Andersen JF. Functional and structural similarities of D7 proteins in the independently-evolved salivary secretions of sand flies and mosquitoes. Sci Rep 2019; 9:5340. [PMID: 30926880 PMCID: PMC6440969 DOI: 10.1038/s41598-019-41848-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/18/2019] [Indexed: 11/09/2022] Open
Abstract
The habit of blood feeding evolved independently in many insect orders of families. Sand flies and mosquitoes belong to separate lineages of blood-feeding Diptera and are thus considered to have evolved the trait independently. Because of this, sand fly salivary proteins differ structurally from those of mosquitoes, and orthologous groups are nearly impossible to define. An exception is the long-form D7-like proteins that show conservation with their mosquito counterparts of numerous residues associated with the N-terminal domain binding pocket. In mosquitoes, this pocket is responsible for the scavenging of proinflammatory cysteinyl leukotrienes and thromboxanes at the feeding site. Here we show that long-form D7 proteins AGE83092 and ABI15936 from the sand fly species, Phlebotomus papatasi and P. duboscqi, respectively, inhibit the activation of platelets by collagen and the thromboxane A2 analog U46619. Using isothermal titration calorimetry, we also demonstrate direct binding of U46619 and cysteinyl leukotrienes C4, D4 and E4 to the P. papatasi protein. The crystal structure of P. duboscqi ABI15936 was determined and found to contain two domains oriented similarly to those of the mosquito proteins. The N-terminal domain contains an apparent eicosanoid binding pocket. The C-terminal domain is smaller in overall size than in the mosquito D7s and is missing some helical elements. Consequently, it does not contain an obvious internal binding pocket for small-molecule ligands that bind to many mosquito D7s. Structural similarities indicate that mosquito and sand fly D7 proteins have evolved from similar progenitors, but phylogenetics and differences in intron/exon structure suggest that they may have acquired the ability to bind vertebrate eicosanoids independently, indicating a convergent evolution scenario.
Collapse
Affiliation(s)
- Willy Jablonka
- The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Il Hwan Kim
- The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Patricia H Alvarenga
- The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Jesus G Valenzuela
- The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Jose M C Ribeiro
- The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - John F Andersen
- The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland, 20852, USA.
| |
Collapse
|
25
|
Ribeiro PA, Dias DS, Lage DP, Martins VT, Costa LE, Santos TT, Ramos FF, Tavares GS, Mendonça DV, Ludolf F, Gomes DA, Rodrigues MA, Chávez-Fumagalli MA, Silva ES, Galdino AS, Duarte MC, Roatt BM, Menezes-Souza D, Teixeira AL, Coelho EA. Immunogenicity and protective efficacy of a new Leishmania hypothetical protein applied as a DNA vaccine or in a recombinant form against Leishmania infantum infection. Mol Immunol 2019; 106:108-118. [DOI: 10.1016/j.molimm.2018.12.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 09/01/2018] [Accepted: 12/21/2018] [Indexed: 01/02/2023]
|
26
|
Manning JE, Cantaert T. Time to Micromanage the Pathogen-Host-Vector Interface: Considerations for Vaccine Development. Vaccines (Basel) 2019; 7:E10. [PMID: 30669682 PMCID: PMC6466432 DOI: 10.3390/vaccines7010010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/10/2019] [Accepted: 01/16/2019] [Indexed: 12/18/2022] Open
Abstract
The current increase in vector-borne disease worldwide necessitates novel approaches to vaccine development targeted to pathogens delivered by blood-feeding arthropod vectors into the host skin. A concept that is gaining traction in recent years is the contribution of the vector or vector-derived components, like salivary proteins, to host-pathogen interactions. Indeed, the triad of vector-host-pathogen interactions in the skin microenvironment can influence host innate and adaptive responses alike, providing an advantage to the pathogen to establish infection. A better understanding of this "bite site" microenvironment, along with how host and vector local microbiomes immunomodulate responses to pathogens, is required for future vaccines for vector-borne diseases. Microneedle administration of such vaccines may more closely mimic vector deposition of pathogen and saliva into the skin with the added benefit of near painless vaccine delivery. Focusing on the 'micro'⁻from microenvironments to microbiomes to microneedles⁻may yield an improved generation of vector-borne disease vaccines in today's increasingly complex world.
Collapse
Affiliation(s)
- Jessica E Manning
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh 12201, Cambodia.
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh 12201, Cambodia.
| |
Collapse
|
27
|
Manning JE, Oliveira F, Parker DM, Amaratunga C, Kong D, Man S, Sreng S, Lay S, Nang K, Kimsan S, Sokha L, Kamhawi S, Fay MP, Suon S, Ruhl P, Ackerman H, Huy R, Wellems TE, Valenzuela JG, Leang R. The PAGODAS protocol: pediatric assessment group of dengue and Aedes saliva protocol to investigate vector-borne determinants of Aedes-transmitted arboviral infections in Cambodia. Parasit Vectors 2018; 11:664. [PMID: 30572920 PMCID: PMC6300895 DOI: 10.1186/s13071-018-3224-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/20/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Mosquito-borne arboviruses, like dengue virus, continue to cause significant global morbidity and mortality, particularly in Southeast Asia. When the infectious mosquitoes probe into human skin for a blood meal, they deposit saliva containing a myriad of pharmacologically active compounds, some of which alter the immune response and influence host receptivity to infection, and consequently, the establishment of the virus. Previous reports have highlighted the complexity of mosquito vector-derived factors and immunity in the success of infection. Cumulative evidence from animal models and limited data from humans have identified various vector-derived components, including salivary components, that are co-delivered with the pathogen and play an important role in the dissemination of infection. Much about the roles and effects of these vector-derived factors remain to be discovered. METHODS/DESIGN We describe a longitudinal, pagoda (community)-based pediatric cohort study to evaluate the burden of dengue virus infection and document the immune responses to salivary proteins of Aedes aegypti, the mosquito vector of dengue, Zika, and chikungunya viruses. The study includes community-based seroprevalence assessments in the peri-urban town of Chbar Mon in Kampong Speu Province, Cambodia. The study aims to recruit 771 children between the ages of 2 and 9 years for a three year period of longitudinal follow-up, including twice per year (rainy and dry season) serosurveillance for dengue seroconversion and Ae. aegypti salivary gland homogenate antibody intensity determinations by ELISA assays. Diagnostic tests for acute dengue, Zika and chikungunya viral infections will be performed by RT-PCR. DISCUSSION This study will serve as a foundation for further understanding of mosquito saliva immunity and its impact on Aedes-transmitted arboviral diseases endemic to Cambodia. TRIAL REGISTRATION NCT03534245 registered on 23 May 2018.
Collapse
Affiliation(s)
- Jessica E. Manning
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland USA
| | - Fabiano Oliveira
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland USA
| | - Daniel M. Parker
- Department of Population Health and Disease Prevention, University of California, Irvine, California, USA
| | - Chanaki Amaratunga
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland USA
| | - Dara Kong
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Somnang Man
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Sokunthea Sreng
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Sreyngim Lay
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Kimsour Nang
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Soun Kimsan
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Ly Sokha
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Shaden Kamhawi
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland USA
| | - Michael P. Fay
- Biostatistics Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland USA
| | - Seila Suon
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Parker Ruhl
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland USA
| | - Hans Ackerman
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland USA
| | - Rekol Huy
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Thomas E. Wellems
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland USA
| | - Jesus G. Valenzuela
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland USA
| | - Rithea Leang
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| |
Collapse
|
28
|
High-through identification of T cell-specific phage-exposed mimotopes using PBMCs from tegumentary leishmaniasis patients and their use as vaccine candidates against Leishmania amazonensis infection. Parasitology 2018; 146:322-332. [PMID: 30198459 DOI: 10.1017/s0031182018001403] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In the current study, phage-exposed mimotopes as targets against tegumentary leishmaniasis (TL) were selected by means of bio-panning cycles employing sera of TL patients and healthy subjects, besides the immune stimulation of peripheral blood mononuclear cells (PBMCs) collected from untreated and treated TL patients and healthy subjects. The clones were evaluated regarding their specific interferon-γ (IFN-γ) and interleukin-4 (IL-4) production in the in vitro cultures, and selectivity and specificity values were calculated, and those presenting the best results were selected for the in vivo experiments. Two clones, namely A4 and A8, were identified and used in immunization protocols from BALB/c mice to protect against Leishmania amazonensis infection. Results showed a polarized Th1 response generated after vaccination, being based on significantly higher levels of IFN-γ, IL-2, IL-12, tumour necrosis factor-α (TNF-α) and granulocyte-macrophage colony-stimulating factor (GM-CSF); which were associated with lower production of specific IL-4, IL-10 and immunoglobulin G1 (IgG1) antibodies. Vaccinated mice presented significant reductions in the parasite load in the infected tissue and distinct organs, when compared with controls. In conclusion, we presented a strategy to identify new mimotopes able to induce Th1 response in PBMCs from TL patients and healthy subjects, and that were successfully used to protect against L. amazonensis infection.
Collapse
|
29
|
Vijayakumar S, Das P. Recent progress in drug targets and inhibitors towards combating leishmaniasis. Acta Trop 2018; 181:95-104. [PMID: 29452111 DOI: 10.1016/j.actatropica.2018.02.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/24/2018] [Accepted: 02/11/2018] [Indexed: 12/22/2022]
Abstract
Lesihmaniasis is one of the major neglected tropical disease caused by the parasite of the genus Leishmania. The disease has more than one clinical forms and the visceral form is considered fatal. With the lack of potential vaccine, chemotherapy is the major treatment source considered for the control of the disease in the infected people. Drugs including amphotericin B and miltefosine are widely used for the treatment, however, development of resistance by the parasite towards the administered drug and high-toxicity of the drug are of major concern. Hence, more attention has been shown on identifying new targets, effective inhibitors, and better drug delivery system against the disease. This review deals with recent studies on drug targets and exploring their essentiality for the survival of Leishmania. Further, new inhibitors for those targets, novel anti-leishmanial peptides and vaccines against leishmaniasis were discussed. We believe that this pool of information will ease the researchers to gain knowledge and help in choosing right targets and design of new inhibitors against Leishmaniasis.
Collapse
|
30
|
Human Interleukin-32γ Plays a Protective Role in an Experimental Model of Visceral Leishmaniasis in Mice. Infect Immun 2018; 86:IAI.00796-17. [PMID: 29483288 DOI: 10.1128/iai.00796-17] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/03/2018] [Indexed: 12/17/2022] Open
Abstract
Visceral leishmaniasis (VL) is a chronic parasitic disease caused by Leishmania infantum in the Americas. During VL, several proinflammatory cytokines are produced in spleen, liver, and bone marrow. However, the role of interleukin-32 (IL-32) has not been explored in this disease. IL-32 can induce production of proinflammatory cytokines in innate immune cells and polarize the adaptive immune response. Herein, we discovered that L. infantum antigens induced expression of mRNA mainly for the IL-32γ isoform but also induced low levels of the IL-32β transcript in human peripheral blood mononuclear cells. Furthermore, infection of human IL-32γ transgenic mice (IL-32γTg mice) with L. infantum promastigote forms increased IL-32γ expression in the spleen and liver. Interestingly, IL-32γTg mice harbored less parasitism in the spleen and liver than wild-type (WT) mice. In addition, IL-32γTg mice showed increased granuloma formation in the liver compared to WT mice. The protection against VL was associated with increased production of nitric oxide (NO), interferon gamma (IFN-γ), IL-17A, and tumor necrosis factor alpha by splenic cells restimulated ex vivo with L. infantum antigens. In parallel, there was an increase in the number of Th1 and Th17 T cells in the spleens of IL-32γTg mice infected with L. infantum IL-32γ induction of IFN-γ and IL-17A expression was found to be essential for NO production by splenic cells of infected animals. These data indicate that IL-32γ potentiates the Th1/Th17 immune response during experimental VL, thus contributing to the control of L. infantum infection.
Collapse
|
31
|
Ghorbani M, Farhoudi R. Leishmaniasis in humans: drug or vaccine therapy? DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 12:25-40. [PMID: 29317800 PMCID: PMC5743117 DOI: 10.2147/dddt.s146521] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Leishmania is an obligate intracellular pathogen that invades phagocytic host cells. Approximately 30 different species of Phlebotomine sand flies can transmit this parasite either anthroponotically or zoonotically through their bites. Leishmaniasis affects poor people living around the Mediterranean Basin, East Africa, the Americas, and Southeast Asia. Affected regions are often remote and unstable, with limited resources for treating this disease. Leishmaniasis has been reported as one of the most dangerous neglected tropical diseases, second only to malaria in parasitic causes of death. People can carry some species of Leishmania for long periods without becoming ill, and symptoms depend on the form of the disease. There are many drugs and candidate vaccines available to treat leishmaniasis. For instance, antiparasitic drugs, such as amphotericin B (AmBisome), are a treatment of choice for leishmaniasis depending on the type of the disease. Despite the availability of different treatment approaches to treat leishmaniasis, therapeutic tools are not adequate to eradicate this infection. In the meantime, drug therapy has been limited because of adverse side effects and unsuccessful vaccine preparation. However, it can immediately make infections inactive. According to other studies, vaccination cannot eradicate leishmaniasis. There is no perfect vaccine or suitable drug to eradicate leishmaniasis completely. So far, no vaccine or drug has been provided to induce long-term protection and ensure effective immunity against leishmaniasis. Therefore, it is necessary that intensive research should be performed in drug and vaccine fields to achieve certain results.
Collapse
Affiliation(s)
- Masoud Ghorbani
- Department of Viral Vaccine Production, Pasteur Institute of Iran, Research and Production Complex, Karaj, Iran
| | - Ramin Farhoudi
- Department of Viral Vaccine Production, Pasteur Institute of Iran, Research and Production Complex, Karaj, Iran
| |
Collapse
|
32
|
Antigenicity, immunogenicity and protective efficacy of a conserved Leishmania hypothetical protein against visceral leishmaniasis. Parasitology 2017; 145:740-751. [PMID: 29113597 DOI: 10.1017/s0031182017001731] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In this study, a Leishmania hypothetical protein, LiHyS, was evaluated regarding its antigenicity, immunogenicity and protective efficacy against visceral leishmaniasis (VL). Regarding antigenicity, immunoblottings and an enzyme-linked immunosorbent assay using human and canine sera showed high sensitivity and specificity values for the recombinant protein (rLiHyS) in the diagnosis of VL. When evaluating the immunogenicity of LiHyS, which is possibly located in the parasite's flagellar pocket, proliferative assays using peripheral blood mononuclear cells from healthy subjects or VL patients showed a high proliferative index in both individuals, when compared to the results obtained using rA2 or unstimulated cultures. Later, rLiHyS/saponin was inoculated in BALB/c mice, which were then challenged with Leishmania infantum promastigotes. The vaccine induced an interferon-γ, interleukin (IL)-12 and granulocyte-macrophage colony-stimulating factor production, which was maintained after infection and which was associated with high nitrite and IgG2a antibody levels, as well as low IL-4 and IL-10 production. Significant reductions in the parasite load in liver, spleen, bone marrow and draining lymph nodes were found in these animals. In this context, the present study shows that the rLiHyS has the capacity to be evaluated as a diagnostic marker or vaccine candidate against VL.
Collapse
|
33
|
Iniguez E, Schocker NS, Subramaniam K, Portillo S, Montoya AL, Al-Salem WS, Torres CL, Rodriguez F, Moreira OC, Acosta-Serrano A, Michael K, Almeida IC, Maldonado RA. An α-Gal-containing neoglycoprotein-based vaccine partially protects against murine cutaneous leishmaniasis caused by Leishmania major. PLoS Negl Trop Dis 2017; 11:e0006039. [PMID: 29069089 PMCID: PMC5673233 DOI: 10.1371/journal.pntd.0006039] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/06/2017] [Accepted: 10/15/2017] [Indexed: 11/22/2022] Open
Abstract
Background Protozoan parasites from the genus Leishmania cause broad clinical manifestations known as leishmaniases, which affect millions of people worldwide. Cutaneous leishmaniasis (CL), caused by L. major, is one the most common forms of the disease in the Old World. There is no preventive or therapeutic human vaccine available for L. major CL, and existing drug treatments are expensive, have toxic side effects, and resistant parasite strains have been reported. Hence, further therapeutic interventions against the disease are necessary. Terminal, non-reducing, and linear α-galactopyranosyl (α-Gal) epitopes are abundantly found on the plasma membrane glycolipids of L. major known as glycoinositolphospholipids. The absence of these α-Gal epitopes in human cells makes these glycans highly immunogenic and thus potential targets for vaccine development against CL. Methodology/Principal findings Here, we evaluated three neoglycoproteins (NGPs), containing synthetic α-Gal epitopes covalently attached to bovine serum albumin (BSA), as vaccine candidates against L. major, using α1,3-galactosyltransferase-knockout (α1,3GalT-KO) mice. These transgenic mice, similarly to humans, do not express nonreducing, linear α-Gal epitopes in their cells and are, therefore, capable of producing high levels of anti-α-Gal antibodies. We observed that Galα(1,6)Galβ-BSA (NGP5B), but not Galα(1,4)Galβ-BSA (NGP12B) or Galα(1,3)Galα-BSA (NGP17B), was able to significantly reduce the size of footpad lesions by 96% in comparison to control groups. Furthermore, we observed a robust humoral and cellular immune response with production of high levels of protective lytic anti-α-Gal antibodies and induction of Th1 cytokines. Conclusions/Significance We propose that NGP5B is an attractive candidate for the study of potential synthetic α-Gal-neoglycoprotein-based vaccines against L. major infection. Despite a worldwide prevalence, cutaneous leishmaniasis (CL) remains largely neglected, with no prophylactic or therapeutic vaccine available. In the Old World, CL is mainly caused by either Leishmania major or L. tropica parasites, which produce localized cutaneous ulcers, often leading to scarring and social stigma. Currently, the disease has reached hyperendemicity levels in the Middle East due to conflict and human displacement. Furthermore, the first choice of treatment in that region continues to be pentavalent antimonials, which are costly and highly toxic, and current vector control measures alone are not sufficient to stop disease transmission. Hence, a vaccine against CL would be very beneficial. Previous studies have demonstrated that sugars are promising vaccine candidates against leishmaniasis, since most parasite species have a cell surface coat composed of immunogenic sugars, including linear α-galactopyranosyl (α-Gal) epitopes, which are absent in humans. Here, we have developed an α-Gal-based vaccine candidate, named NGP5B. When tested in transgenic mice which like humans lack α-Gal epitopes in their cells, NGP5B was able to induce a significant partial protection against L. major infection, by significantly reducing mouse footpad lesions and parasite burden. Altogether, we propose NGP5B as a promising preventive vaccine for CL caused by L. major.
Collapse
Affiliation(s)
- Eva Iniguez
- Department of Biological Sciences, Border Biomedical Research Center, the University of Texas at El Paso, El Paso, Texas, United States of America
| | - Nathaniel S. Schocker
- Department of Chemistry, Border Biomedical Research Center, the University of Texas at El Paso, El Paso, Texas, United States of America
| | - Krishanthi Subramaniam
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Susana Portillo
- Department of Biological Sciences, Border Biomedical Research Center, the University of Texas at El Paso, El Paso, Texas, United States of America
| | - Alba L. Montoya
- Department of Chemistry, Border Biomedical Research Center, the University of Texas at El Paso, El Paso, Texas, United States of America
| | - Waleed S. Al-Salem
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Caresse L. Torres
- Department of Biological Sciences, Border Biomedical Research Center, the University of Texas at El Paso, El Paso, Texas, United States of America
| | - Felipe Rodriguez
- Department of Biological Sciences, Border Biomedical Research Center, the University of Texas at El Paso, El Paso, Texas, United States of America
| | - Otacilio C. Moreira
- Laboratório de Biologia Molecular e Doenças Endêmicas, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alvaro Acosta-Serrano
- Department of Parasitology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, United Kingdom
| | - Katja Michael
- Department of Chemistry, Border Biomedical Research Center, the University of Texas at El Paso, El Paso, Texas, United States of America
| | - Igor C. Almeida
- Department of Biological Sciences, Border Biomedical Research Center, the University of Texas at El Paso, El Paso, Texas, United States of America
- * E-mail: (ICA); (RAM)
| | - Rosa A. Maldonado
- Department of Biological Sciences, Border Biomedical Research Center, the University of Texas at El Paso, El Paso, Texas, United States of America
- * E-mail: (ICA); (RAM)
| |
Collapse
|
34
|
Wheat WH, Arthun EN, Spencer JS, Regan DP, Titus RG, Dow SW. Immunization against full-length protein and peptides from the Lutzomyia longipalpis sand fly salivary component maxadilan protects against Leishmania major infection in a murine model. Vaccine 2017; 35:6611-6619. [PMID: 29079105 PMCID: PMC5710984 DOI: 10.1016/j.vaccine.2017.10.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 10/02/2017] [Accepted: 10/13/2017] [Indexed: 02/06/2023]
Abstract
Leishmaniasis is an arthropod vectored disease causing considerable human morbidity and mortality. Vaccination remains the most realistic and practical means to interrupt the growing number and diversity of sand fly vectors and reservoirs of Leishmania. Since transmission of Leishmania is achieved exclusively by sand fly vectors via immune-modulating salivary substances, conventional vaccination requiring an unmodified host immune response for success are potentially destined to fail unless immunomodulatory factors are somehow neutralized. Using cationic liposome DNA complexes (CLDC) as an adjuvant system along with Lu. longipalpis sand fly salivary component maxadilan (MAX) as antigen (Ag), we show that mice are protected from the MAX-induced exacerbation of infection with Leishmania major (Lm). The CLDC adjuvant and alum were comparable in terms of lesion induration and decreased parasite burden, however the alum adjuvant imposed more inflammation at the injection site. BALB/c, C3H and C57BL/6 mice vaccinated with MAX-CLDC containing either the full-length MAX or peptides spanning the N- and C-terminal regions of MAX are protected against footpad challenges with Lm co-injected with MAX. When compared to unvaccinated controls, all strains of mice immunized with CLDC containing either peptides encompassing the first 20 N-terminal AA or those spanning the last 15 AA of the C-terminal domain of MAX demonstrated decreased parasite burden after 9 or 18 weeks post challenge with Lm + MAX. MAX-CLDC immunized mice showed increased IFNγ-secreting and decreased IL-4-secreting CD4+ cells in footpad-draining lymph nodes. Antisera from C-terminal peptide (P11) MAX-CLDC-vaccinated animals was capable of recognizing FL-MAX and its C-terminal domain and also blocked MAX-mediated reprogramming of bone marrow-derived dendritic cells (BM-DC) in vitro. This peptide vaccine targeting sand fly MAX, improves host immunity against MAX-mediated immunomodulation.
Collapse
Affiliation(s)
- William H Wheat
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, United States; Department of Microbiology, Immunology, Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, United States.
| | - Erik N Arthun
- Department of Biology, College of Natural Sciences, Colorado State University, Fort Collins, CO 80523, United States
| | - John S Spencer
- Department of Microbiology, Immunology, Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, United States
| | - Daniel P Regan
- Department of Microbiology, Immunology, Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, United States
| | - Richard G Titus
- Department of Microbiology, Immunology, Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, United States
| | - Steven W Dow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, United States; Department of Microbiology, Immunology, Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, United States
| |
Collapse
|
35
|
Tlili A, Marzouki S, Chabaane E, Abdeladhim M, Kammoun-Rebai W, Sakkouhi R, Belhadj Hmida N, Oliveira F, Kamhawi S, Louzir H, Valenzuela JG, Ben Ahmed M. Phlebotomus papatasi Yellow-Related and Apyrase Salivary Proteins Are Candidates for Vaccination against Human Cutaneous Leishmaniasis. J Invest Dermatol 2017; 138:598-606. [PMID: 29054598 DOI: 10.1016/j.jid.2017.09.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/25/2017] [Accepted: 09/06/2017] [Indexed: 10/18/2022]
Abstract
Nowadays, there is no available vaccine for human leishmaniasis. Animal experiments demonstrate that pre-exposure to sand fly saliva confers protection against leishmaniasis. Our preceding work in humans indicates that Phlebotomus papatasi saliva induces the production of IL-10 by CD8+ T lymphocytes. The neutralization of IL-10 enhanced the activation of a T-cell CD4+ population-producing IFN-γ. Herein, we used a biochemical and functional genomics approach to identify the sand fly salivary components that are responsible for the activation of the T helper type 1 immune response in humans, therefore constituting potential vaccine candidates against leishmaniasis. Fractionated P. papatasi salivary extracts were first tested on T lymphocytes of immune donors. We confirmed that the CD4+ lymphocytes proliferate and produce IFN-γ in response to stimulation with the proteins of molecular weight >30 kDa. Peripheral blood mononuclear cells from immune donors were transfected with plasmids coding for the most abundant proteins from the P. papatasi salivary gland cDNA library. Our result showed that the "yellow related proteins," PPTSP42 and PPTSP44, and "apyrase," PPTSP36, are the proteins responsible for the aforementioned cellular immune response and IFN-γ production. Strikingly, PPTSP44 triggered the highest level of lymphocyte proliferation and IFN-γ production. Multiplex cytokine analysis confirmed the T helper type 1-polarized response induced by these proteins. Importantly, recombinant PPTSP44 validated the results observed with the DNA plasmid, further supporting that PPTSP44 constitutes a promising vaccine candidate against human leishmaniasis.
Collapse
Affiliation(s)
- Aymen Tlili
- Laboratory of Transmission, Control and Immunobiology of Infections, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Soumaya Marzouki
- Laboratory of Transmission, Control and Immunobiology of Infections, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Emna Chabaane
- Laboratory of Transmission, Control and Immunobiology of Infections, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Maha Abdeladhim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Wafa Kammoun-Rebai
- Laboratory of Medical Parasitology, Biotechnologies and Biomolecules, LR11IPT06, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Rahma Sakkouhi
- Laboratory of Transmission, Control and Immunobiology of Infections, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Nabil Belhadj Hmida
- Laboratory of Transmission, Control and Immunobiology of Infections, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Hechmi Louzir
- Laboratory of Transmission, Control and Immunobiology of Infections, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia; Faculté de Médecine de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - 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, Maryland, USA
| | - Mélika Ben Ahmed
- Laboratory of Transmission, Control and Immunobiology of Infections, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia; Faculté de Médecine de Tunis, Université Tunis El Manar, Tunis, Tunisia.
| |
Collapse
|
36
|
Kammoun-Rebai W, Bahi-Jaber N, Naouar I, Toumi A, Ben Salah A, Louzir H, Meddeb-Garnaoui A. Human cellular and humoral immune responses to Phlebotomus papatasi salivary gland antigens in endemic areas differing in prevalence of Leishmania major infection. PLoS Negl Trop Dis 2017; 11:e0005905. [PMID: 29023574 PMCID: PMC5638224 DOI: 10.1371/journal.pntd.0005905] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 08/24/2017] [Indexed: 01/22/2023] Open
Abstract
Background Sand fly saliva compounds are able to elicit specific immune responses that have a significant role in Leishmania parasite establishment and disease outcome. Characterizing anti-saliva immune responses in individuals living in well defined leishmaniasis endemic areas would provide valuable insights regarding their effect on parasite transmission and establishment in humans. Methodology/Principal findings We explored the cellular and humoral immune responses to Phlebotomus (P.) papatasi salivary gland extracts (SGE) in individuals living in cutaneous leishmaniasis (CL) old or emerging foci (OF, EF). OF was characterized by a higher infection prevalence as assessed by higher proportions of leishmanin skin test (LST) positive individuals compared to EF. Subjects were further subdivided into healed, asymptomatic or naïve groups. We showed anti-SGE proliferation in less than 30% of the individuals, regardless of the immune status, in both foci. IFN-γ production was higher in OF and only observed in immune individuals from OF and naïve subjects from EF. Although IL-10 was not detected, addition of anti-human IL-10 antibodies revealed an increase in proliferation and IFN-γ production only in individuals from OF. The percentage of seropositive individuals was similar in immune and naïves groups but was significantly higher in OF. No correlation was observed between anti-saliva immune responses and LST response. High anti-SGE-IgG responses were associated with an increased risk of developing ZCL. No differences were observed for anti-SGE humoral or cellular responses among naïve individuals who converted or not their LST response or developed or not ZCL after the transmission season. Conclusions/Significance These data suggest that individuals living in an old focus characterized by a frequent exposure to sand fly bites and a high prevalence of infection, develop higher anti-saliva IgG responses and IFN-γ levels and a skew towards a Th2-type cellular response, probably in favor of parasite establishment, compared to those living in an emerging focus. During murine experimental leishmaniasis sand fly saliva components modulate the host immune response and facilitate infection while pre-exposition to uninfected sand fly bites is associated with a protective cellular response against subsequent infection. Human anti-saliva immune responses are not well defined in leishmaniasis endemic areas. Here, we report an analysis of anti P. papatasi saliva cellular and humoral responses in individuals residing in endemic foci showing different prevalence rates of L. major infection. Individuals were further subdivided based on LST response and presence of typical CL scars. We showed higher anti-saliva cellular and humoral responses and a skew towards a Th2 response in the old focus characterized by the highest prevalence of infection. No correlation was observed between LST and anti-saliva cellular or humoral response. We showed that high anti-saliva IgG responses constituted a risk factor for the development of CL. Our findings suggest that the anti-P. papatasi saliva cellular and humoral response profiles vary with the level of sand fly exposure and the prevalence of infection in CL endemic areas. Such studies in humans from highly endemic areas could contribute to a better understanding of the immune response to sand fly saliva and its role in leishmaniasis outcome.
Collapse
Affiliation(s)
- Wafa Kammoun-Rebai
- Laboratory of Medical Parasitology, Biotechnologies and Biomolecules, Institut Pasteur de Tunis, Tunis, Tunisia
- Département de Biologie, Université Tunis El Manar, Tunis, Tunisia
| | - Narges Bahi-Jaber
- UPSP EGEAL Institut Polytechnique LaSalle Beauvais, Beauvais, France
| | - Ikbel Naouar
- Département de Biologie, Université Tunis El Manar, Tunis, Tunisia
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Amine Toumi
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Afif Ben Salah
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunis, Tunisia
- Arabian Gulf University, College of Medicine and Medical Sciences, Manama, Bahrain
| | - Hechmi Louzir
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunis, Tunisia
- Faculté de Médecine de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Amel Meddeb-Garnaoui
- Laboratory of Medical Parasitology, Biotechnologies and Biomolecules, Institut Pasteur de Tunis, Tunis, Tunisia
- * E-mail:
| |
Collapse
|
37
|
In silico analysis and in vitro evaluation of immunogenic and immunomodulatory properties of promiscuous peptides derived from Leishmania infantum eukaryotic initiation factor. Bioorg Med Chem 2017; 25:5904-5916. [PMID: 28974324 DOI: 10.1016/j.bmc.2017.07.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 06/16/2017] [Accepted: 07/06/2017] [Indexed: 12/16/2022]
Abstract
It is generally considered as imperative the ability to control leishmaniasis through the development of a protective vaccine capable of inducing long-lasting and protective cell-mediated immune responses. In this current study, we demonstrated potential epitopes that bind to H2 MHC class I and II molecules by conducting the in silico analysis of Leishmania infantum eukaryotic Initiation Factor (LieIF) protein, using online available algorithms. Moreover, we synthesized five peptides (16-18 amino acids long) which are part of the N-terminal portion of LieIF and contain promising MHC class I and II-restricted epitopes and afterwards, their predicted immunogenicity was evaluated in vitro by monitoring peptide-specific T-cell responses. Additionally, the immunomodulatory properties of these peptides were investigated in vitro by exploring their potential of inducing phenotypic maturation and functional differentiation of murine Bone-Marrow derived Dendritic Cells (BM-DCs). It was revealed by our data that all the synthetic peptides predicted for H2 alleles; present the property of immunogenicity. Among the synthetic peptides which contained T-cell epitopes, the peptide 52-68 aa (LieIF_2) exhibited immunomodulatory properties with the larger potential. LieIF_2-pulsed BM-DCs up-regulated the expression of the co-stimulatory surface molecules CD80 and CD86, as well as the production of the proinflammatory cytokine TNF-α and of the Th1-polarizing cytokines IL-12 and IFN-γ. The aforementioned data suggest that selected parts of LieIF could be used to develop innovative subunit protective vaccines able to induce effective immunity mediated by MHC class I-restricted as well as class II-restricted T-cell responses.
Collapse
|
38
|
Probing the efficacy of a heterologous Leishmania/L. Viannia braziliensis recombinant enolase as a candidate vaccine to restrict the development of L. infantum in BALB/c mice. Acta Trop 2017; 171:8-16. [PMID: 28288798 DOI: 10.1016/j.actatropica.2017.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/11/2017] [Accepted: 03/09/2017] [Indexed: 11/23/2022]
Abstract
In the present study, the Leishmania braziliensis enolase protein was evaluated as a vaccine candidate against visceral leishmaniasis (VL). The DNA sequence was cloned and the recombinant protein (rEnolase) was evaluated as a vaccine, associated with saponin, as an immune adjuvant. The protective efficacy of the rEnolase plus saponin combination was investigated in BALB/c mice against Leishmania infantum infection. The results revealed that the vaccine induced higher levels of IFN-γ, IL-12, and GM-CSF when a capture ELISA and flow cytometry were performed, as well as an antileishmanial nitrite production after using in vitro stimulation with rEnolase and an antigenic Leishmania preparation. The vaccinated animals, when compared to the control groups, showed a lower parasite burden in the liver, spleen, bone marrow, and paws' draining lymph nodes when both a limiting dilution technique and RT-PCR assay were performed. In addition, these mice showed low levels of antileishmanial IL-4, IL-10, and anti-Leishmania IgG1 isotype antibodies. Partial protection was associated with IFN-γ production, which was mainly mediated by CD4+ T cells. In conclusion, the present study's data showed that the L. braziliensis enolase protein could be considered a vaccine candidate that offers heterologous protection against VL.
Collapse
|
39
|
|
40
|
A recombinant fusion protein displaying murine and human MHC class I- and II-specific epitopes protects against Leishmania amazonensis infection. Cell Immunol 2017; 313:32-42. [DOI: 10.1016/j.cellimm.2016.12.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/24/2016] [Accepted: 12/25/2016] [Indexed: 12/21/2022]
|
41
|
Duarte MC, Lage DP, Martins VT, Chávez-Fumagalli MA, Roatt BM, Menezes-Souza D, Goulart LR, Soto M, Tavares CAP, Coelho EAF. Recent updates and perspectives on approaches for the development of vaccines against visceral leishmaniasis. Rev Soc Bras Med Trop 2017; 49:398-407. [PMID: 27598624 DOI: 10.1590/0037-8682-0120-2016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 06/09/2016] [Indexed: 11/22/2022] Open
Abstract
Visceral leishmaniasis (VL) is one of the most important tropical diseases worldwide. Although chemotherapy has been widely used to treat this disease, problems related to the development of parasite resistance and side effects associated with the compounds used have been noted. Hence, alternative approaches for VL control are desirable. Some methods, such as vector control and culling of infected dogs, are insufficiently effective, with the latter not ethically recommended. The development of vaccines to prevent VL is a feasible and desirable measure for disease control; for example, some vaccines designed to protect dogs against VL have recently been brought to market. These vaccines are based on the combination of parasite fractions or recombinant proteins with adjuvants that are able to induce cellular immune responses; however, their partial efficacy and the absence of a vaccine to protect against human leishmaniasis underline the need for characterization of new vaccine candidates. This review presents recent advances in control measures for VL based on vaccine development, describing extensively studied antigens, as well as new antigenic proteins recently identified using immuno-proteomic techniques.
Collapse
Affiliation(s)
- Mariana Costa Duarte
- Departamento de Patologia Clínica, Colégio Técnico, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniela Pagliara Lage
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vívian Tamietti Martins
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Miguel Angel Chávez-Fumagalli
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bruno Mendes Roatt
- Departamento de Patologia Clínica, Colégio Técnico, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniel Menezes-Souza
- Departamento de Patologia Clínica, Colégio Técnico, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luiz Ricardo Goulart
- Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, Uberlândia, Minas Gerais, Brazil.,Department of Medical Microbiology and Immunology, University of California-Davis, Davis, CA, USA
| | - Manuel Soto
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlos Alberto Pereira Tavares
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Eduardo Antonio Ferraz Coelho
- Departamento de Patologia Clínica, Colégio Técnico, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
42
|
Martins VT, Duarte MC, Lage DP, Costa LE, Carvalho AMRS, Mendes TAO, Roatt BM, Menezes-Souza D, Soto M, Coelho EAF. A recombinant chimeric protein composed of human and mice-specific CD4+and CD8+T-cell epitopes protects against visceral leishmaniasis. Parasite Immunol 2016; 39. [DOI: 10.1111/pim.12359] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/30/2016] [Indexed: 12/28/2022]
Affiliation(s)
- V. T. Martins
- Departamento de Bioquímica e Imunologia; Instituto de Ciências Biológicas; Universidade Federal de Minas Gerais; Belo Horizonte Minas Gerais Brazil
| | - M. C. Duarte
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical; Faculdade de Medicina; Universidade Federal de Minas Gerais; Belo Horizonte Minas Gerais Brazil
- Departamento de Patologia Clínica; COLTEC; Universidade Federal de Minas Gerais; Belo Horizonte Minas Gerais Brazil
| | - D. P. Lage
- Departamento de Patologia Clínica; COLTEC; Universidade Federal de Minas Gerais; Belo Horizonte Minas Gerais Brazil
| | - L. E. Costa
- Departamento de Patologia Clínica; COLTEC; Universidade Federal de Minas Gerais; Belo Horizonte Minas Gerais Brazil
| | - A. M. R. S. Carvalho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical; Faculdade de Medicina; Universidade Federal de Minas Gerais; Belo Horizonte Minas Gerais Brazil
| | - T. A. O. Mendes
- Departamento de Bioquímica e Imunologia; Instituto de Ciências Biológicas; Universidade Federal de Minas Gerais; Belo Horizonte Minas Gerais Brazil
| | - B. M. Roatt
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical; Faculdade de Medicina; Universidade Federal de Minas Gerais; Belo Horizonte Minas Gerais Brazil
| | - D. Menezes-Souza
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical; Faculdade de Medicina; Universidade Federal de Minas Gerais; Belo Horizonte Minas Gerais Brazil
- Departamento de Patologia Clínica; COLTEC; Universidade Federal de Minas Gerais; Belo Horizonte Minas Gerais Brazil
| | - M. Soto
- Departamento de Biología Molecular; Centro de Biología Molecular Severo Ochoa; CSIC-UAM; Universidad Autónoma de Madrid; Madrid Spain
| | - E. A. F. Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical; Faculdade de Medicina; Universidade Federal de Minas Gerais; Belo Horizonte Minas Gerais Brazil
- Departamento de Patologia Clínica; COLTEC; Universidade Federal de Minas Gerais; Belo Horizonte Minas Gerais Brazil
| |
Collapse
|
43
|
Jerônimo SMB, Pearson RD. The challenges on developing vaccine against visceral leishmaniasis. Rev Soc Bras Med Trop 2016; 49:395-7. [DOI: 10.1590/0037-8682-0343-2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 08/26/2016] [Indexed: 02/01/2023] Open
Affiliation(s)
- Selma Maria Bezerra Jerônimo
- Universidade Federal do Rio Grande do Norte, Brazil; Universidade Federal do Rio Grande do Norte, Brazil; Instituto Nacional de Ciências e Tecnologia de Doenças Tropicais, Brazil
| | | |
Collapse
|
44
|
Abstract
Cutaneous leishmaniasis is a major public health problem and causes a range of diseases from self-healing infections to chronic disfiguring disease. Currently, there is no vaccine for leishmaniasis, and drug therapy is often ineffective. Since the discovery of CD4(+) T helper 1 (TH1) cells and TH2 cells 30 years ago, studies of cutaneous leishmaniasis in mice have answered basic immunological questions concerning the development and maintenance of CD4(+) T cell subsets. However, new strategies for controlling the human disease have not been forthcoming. Nevertheless, advances in our knowledge of the cells that participate in protection against Leishmania infection and the cells that mediate increased pathology have highlighted new approaches for vaccine development and immunotherapy. In this Review, we discuss the early events associated with infection, the CD4(+) T cells that mediate protective immunity and the pathological role that CD8(+) T cells can have in cutaneous leishmaniasis.
Collapse
|