1
|
Sepahpour T, Alshaweesh J, Azodi N, Singh K, Ireland DDC, Valanezhad F, Nakamura R, Satoskar AR, Dey R, Hamano S, Nakhasi HL, Gannavaram S. Downregulation of IRF7-mediated type-I interferon response by LmCen -/- parasites is necessary for protective immunity. NPJ Vaccines 2024; 9:250. [PMID: 39702382 DOI: 10.1038/s41541-024-01032-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/19/2024] [Indexed: 12/21/2024] Open
Abstract
Leishmaniasis is a tropical disease caused by Leishmania parasites and currently has no licensed vaccines. We developed a dermotropic Leishmania major centrin gene-deleted strain (LmCen-/-) as a live attenuated vaccine. Recent studies have shown that type I interferons (IFNs) play important roles in immunity to parasitic and viral pathogens. However, their relevance in protective immunity following vaccination is not understood. We found that immunization with LmCen-/- induces a transient increase in type I IFN response along with its regulatory factor IRF7 that is downregulated 7-21 days post-immunization, coincided with the induction of a robust Th1 adaptive immune response. Challenge infection with virulent L. donovani parasites showed a significant reduction of splenic and hepatic parasite burden in IRF7-/- mice than wild type mice following immunization with LmCen-/-, suggesting that ablation of type I IFN response is a pre-requisite for the induction of LmCen-/- mediated Th1 immunity against L. donovani infection.
Collapse
Affiliation(s)
- Telly Sepahpour
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, 20993, USA
| | - Jalal Alshaweesh
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), The Joint Usage/Research Center on Tropical Disease, Nagasaki University, Nagasaki, Japan, and Graduate School of Biomedical Sciences, Doctoral Leadership Program, Nagasaki University, Nagasaki, Japan
| | - Nazli Azodi
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, 20993, USA
| | - Komudi Singh
- National Heart Lung Blood Institute (NHLBI), NIH, Bethesda, MD, USA
| | - Derek D C Ireland
- Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Farzaneh Valanezhad
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), The Joint Usage/Research Center on Tropical Disease, Nagasaki University, Nagasaki, Japan, and Graduate School of Biomedical Sciences, Doctoral Leadership Program, Nagasaki University, Nagasaki, Japan
| | - Risa Nakamura
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), The Joint Usage/Research Center on Tropical Disease, Nagasaki University, Nagasaki, Japan, and Graduate School of Biomedical Sciences, Doctoral Leadership Program, Nagasaki University, Nagasaki, Japan
| | - Abhay R Satoskar
- Department of Pathology and Microbiology, Ohio State University, Columbus, OH, USA
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, 20993, USA.
| | - Shinjiro Hamano
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), The Joint Usage/Research Center on Tropical Disease, Nagasaki University, Nagasaki, Japan, and Graduate School of Biomedical Sciences, Doctoral Leadership Program, Nagasaki University, Nagasaki, Japan.
| | - Hira L Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, 20993, USA.
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD, 20993, USA.
| |
Collapse
|
2
|
Webi E, Abkallo HM, Obiero G, Ndegwa P, Xie S, Zhao S, Nene V, Steinaa L. Genome Editing in Apicomplexan Parasites: Current Status, Challenges, and Future Possibilities. CRISPR J 2024; 7:310-326. [PMID: 39387255 DOI: 10.1089/crispr.2024.0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein (Cas) technology has revolutionized genome editing across various biological systems, including the Apicomplexa phylum. This review describes the status, challenges, and applications of CRISPR-Cas9 editing technology in apicomplexan parasites, such as Plasmodium, Toxoplasma, Theileria, Babesia, and Cryptosporidium. The discussion encompasses successfully implemented CRISPR-Cas9-based techniques in these parasites, highlighting the achieved milestones, from precise gene modifications to genome-wide screening. In addition, the review addresses the challenges hampering efficient genome editing, including the parasites' complex life cycles, multiple intracellular stages, and the lack of robust genetic tools. It further explores the ethical and policy considerations surrounding genome editing and the future perspectives of CRISPR-Cas applications in apicomplexan parasites.
Collapse
Affiliation(s)
- Ethel Webi
- Animal and Human Health Program, International Livestock Research Institute, Nairobi, Kenya
- Department of Biochemistry, University of Nairobi, Nairobi, Kenya
| | - Hussein M Abkallo
- Animal and Human Health Program, International Livestock Research Institute, Nairobi, Kenya
| | - George Obiero
- Department of Biochemistry, University of Nairobi, Nairobi, Kenya
| | - Paul Ndegwa
- Department of Biology, University of Nairobi, Nairobi, Kenya
| | - Shengsong Xie
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, P. R. China
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, P. R. China
| | - Vishvanath Nene
- Animal and Human Health Program, International Livestock Research Institute, Nairobi, Kenya
| | - Lucilla Steinaa
- Animal and Human Health Program, International Livestock Research Institute, Nairobi, Kenya
| |
Collapse
|
3
|
Parkash V, Ashwin H, Dey S, Sadlova J, Vojtkova B, Van Bocxlaer K, Wiggins R, Thompson D, Dey NS, Jaffe CL, Schwartz E, Volf P, Lacey CJN, Layton AM, Kaye PM. Safety and reactogenicity of a controlled human infection model of sand fly-transmitted cutaneous leishmaniasis. Nat Med 2024; 30:3150-3162. [PMID: 39095597 PMCID: PMC11564116 DOI: 10.1038/s41591-024-03146-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 06/20/2024] [Indexed: 08/04/2024]
Abstract
The leishmaniases are globally important parasitic diseases for which no human vaccines are currently available. To facilitate vaccine development, we conducted an open-label observational study to establish a controlled human infection model (CHIM) of sand fly-transmitted cutaneous leishmaniasis (CL) caused by Leishmania major. Between 24 January and 12 August 2022, we exposed 14 participants to L. major-infected Phlebotomus duboscqi. The primary objective was to demonstrate effectiveness of lesion development (take rate) and safety (absence of CL lesion at 12 months). Secondary and exploratory objectives included rate of lesion development, parasite load and analysis of local immune responses by immunohistology and spatial transcriptomics. Lesion development was terminated by therapeutic biopsy (between days 14 and 42 after bite) in ten participants with clinically compatible lesions, one of which was not confirmed by parasite detection. We estimated an overall take rate for CL development of 64% (9/14). Two of ten participants had one and one of ten participants had two lesion recurrences 4-8 months after biopsy that were treated successfully with cryotherapy. No severe or serious adverse events were recorded, but as expected, scarring due to a combination of CL and the biopsy procedure was evident. All participants were lesion free at >12-month follow-up. We provide the first comprehensive map of immune cell distribution and cytokine/chemokine expression in human CL lesions, revealing discrete immune niches. This CHIM offers opportunities for vaccine candidate selection based on human efficacy data and for a greater understanding of immune-mediated pathology. ClinicalTrials.gov identifier: NCT04512742 .
Collapse
Affiliation(s)
- Vivak Parkash
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK
| | - Helen Ashwin
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK
| | - Shoumit Dey
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK
| | - Jovana Sadlova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Barbora Vojtkova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Katrien Van Bocxlaer
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK
- Skin Research Centre, Hull York Medical School, York, UK
| | - Rebecca Wiggins
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK
| | - David Thompson
- York and Scarborough Teaching Hospitals NHS Foundation Trust, York, UK
| | - Nidhi Sharma Dey
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK
| | - Charles L Jaffe
- Department of Microbiology and Molecular Genetics, Kuvin Center for the Study of Infectious and Tropical Diseases, IMRIC, The Hebrew University - Hadassah Medical School, Jerusalem, Israel
| | - Eli Schwartz
- Center for Geographic Medicine and Tropical Diseases, Chaim Sheba Medical Center and the School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Charles J N Lacey
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK
| | - Alison M Layton
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK.
- Skin Research Centre, Hull York Medical School, York, UK.
| | - Paul M Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK.
- Skin Research Centre, Hull York Medical School, York, UK.
| |
Collapse
|
4
|
Younis BM, Wiggins R, Khalil EAG, Osman M, Santoro F, Sonnati C, Keding A, Novedrati M, Montesi G, Noureldein A, Elmukashfi ETA, Mustafa AE, Alamin M, Saeed M, Salman K, Suliman AJ, Musa AEA, Layton AM, Lacey CJN, Kaye PM, Musa AM. A randomized, double-blind phase 2b trial to evaluate efficacy of ChAd63-KH for treatment of post kala-azar dermal leishmaniasis. Mol Ther Methods Clin Dev 2024; 32:101310. [PMID: 39253357 PMCID: PMC11381778 DOI: 10.1016/j.omtm.2024.101310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/26/2024] [Indexed: 09/11/2024]
Abstract
In a recent phase 2a clinical trial, the candidate leishmaniasis vaccine ChAd63-KH was shown to be safe and immunogenic in Sudanese patients with post kala-azar dermal leishmaniasis (PKDL). However, its value as a stand-alone therapeutic was unknown. To assess the therapeutic efficacy of ChAd63-KH, we conducted a randomized, double-blind, placebo-controlled phase 2b trial (ClinicalTrials.gov: NCT03969134). Primary outcomes were safety and efficacy (≥90% improvement in clinical disease). Secondary outcomes were change in severity grade and vaccine-induced immune response. 86 participants with uncomplicated PKDL of ≥6 month duration were randomized to receive ChAd63-KH (7.5 × 1010 viral particles, once by the intramuscular route) or placebo. 75 participants (87%) completed the trial as per protocol. No severe or serious adverse events were observed. At day 90 post-vaccination, 6/40 (15%) and 4/35 (11%) participants in the vaccine and placebo groups, respectively, showed ≥90% clinical improvement (risk ratio [RR] 1.31 [95% confidence interval (CI), 0.40-4.28], p = 0.742). There were also no significant differences in PKDL severity grade between study arms. Whole-blood transcriptomic analysis identified transcriptional modules associated with interferon responses and monocyte and dendritic cell activation. Thus, a single vaccination with ChAd63-KH showed no therapeutic efficacy in this subset of Sudanese patients with PKDL.
Collapse
Affiliation(s)
- Brima M Younis
- Department of Clinical Pathology & Immunology, Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan
| | - Rebecca Wiggins
- 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, Khartoum 11111, Sudan
| | - Mohamed Osman
- York Biomedical Research Institute, Hull York Medical School, University of York, Heslington, York YO10 5DD, UK
| | - Francesco Santoro
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Chiara Sonnati
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Ada Keding
- Department of Health Sciences, University of York, Heslington, York YO10 5DD, UK
| | - Maria Novedrati
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Giorgio Montesi
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Ali Noureldein
- Department of Clinical Pathology & Immunology, Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan
| | - Elmukashfi T A Elmukashfi
- Department of Clinical Pathology & Immunology, Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan
| | - Ala Eldin Mustafa
- Department of Clinical Pathology & Immunology, Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan
| | - Mohammed Alamin
- Department of Clinical Pathology & Immunology, Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan
| | - Mohammed Saeed
- Department of Clinical Pathology & Immunology, Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan
| | - Khalid Salman
- Department of Clinical Pathology & Immunology, Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan
| | - Ahmed J Suliman
- Department of Clinical Pathology & Immunology, Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan
| | - Amin E A Musa
- Department of Clinical Pathology & Immunology, Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan
| | - 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, Khartoum 11111, Sudan
| |
Collapse
|
5
|
Avishek K, Beg MA, Vats K, Singh AK, Dey R, Singh KP, Singh RK, Gannavaram S, Ramesh V, Mulla MSA, Bhatnagar U, Singh S, Nakhasi HL, Salotra P, Selvapandiyan A. Manufacturing and preclinical toxicity of GLP grade gene deleted attenuated Leishmania donovani parasite vaccine. Sci Rep 2024; 14:14636. [PMID: 38918456 PMCID: PMC11199483 DOI: 10.1038/s41598-024-64592-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 06/11/2024] [Indexed: 06/27/2024] Open
Abstract
Centrin1 gene deleted Leishmania donovani parasite (LdCen1-/-) was developed and extensively tested experimentally as an intracellular stage-specific attenuated and immunoprotective live parasite vaccine candidate ex vivo using human PBMCs and in vivo in animals. Here we report manufacturing and pre-clinical evaluation of current Good-Laboratory Practice (cGLP) grade LdCen1-/- parasites, as a prerequisite before proceeding with clinical trials. We screened three batches of LdCen1-/- parasites manufactured in bioreactors under cGLP conditions, for their consistency in genetic stability, attenuation, and safety. One such batch was preclinically tested using human PBMCs and animals (hamsters and dogs) for its safety and protective immunogenicity. The immunogenicity of the CGLP grade LdCen1-/- parasites was similar to one grown under laboratory conditions. The cGLP grade LdCen1-/- parasites were found to be safe and non-toxic in hamsters and dogs even at 3 times the anticipated vaccine dose. When PBMCs from healed visceral leishmaniasis (VL) cases were infected with cGLP LdCen1-/-, there was a significant increase in the stimulation of cytokines that contribute to protective responses against VL. This effect, measured by multiplex ELISA, was greater than that observed in PBMCs from healthy individuals. These results suggest that cGLP grade LdCen1-/- manufactured under cGMP complaint conditions can be suitable for future clinical trials.
Collapse
Affiliation(s)
- Kumar Avishek
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India
| | - Mirza A Beg
- Department of Molecular Medicine, Jamia Hamdard, New Delhi, 110062, India
| | - Kavita Vats
- Department of Molecular Medicine, Jamia Hamdard, New Delhi, 110062, India
| | - Avinash Kumar Singh
- Gennova Biopharmaceuticals, Hinjewadi Phase II, Pune, Maharashtra, 411057, India
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Diseases, CBER, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Kamaleshwar P Singh
- Gennova Biopharmaceuticals, Hinjewadi Phase II, Pune, Maharashtra, 411057, India
| | - Rajesh Kumar Singh
- Gennova Biopharmaceuticals, Hinjewadi Phase II, Pune, Maharashtra, 411057, India
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Diseases, CBER, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - V Ramesh
- Department of Dermatology and STD, ESIC Medical College, Faridabad, Haryana, 121001, India
| | | | - Upendra Bhatnagar
- Vimta Laboratories, Cherlapally, Hyderabad, Telangana, 500051, India
| | - Sanjay Singh
- Gennova Biopharmaceuticals, Hinjewadi Phase II, Pune, Maharashtra, 411057, India
| | - Hira L Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, CBER, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Poonam Salotra
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, 110029, India
| | | |
Collapse
|
6
|
Seyed N, Taheri T, Rafati S. Live attenuated-nonpathogenic Leishmania and DNA structures as promising vaccine platforms against leishmaniasis: innovations can make waves. Front Microbiol 2024; 15:1326369. [PMID: 38633699 PMCID: PMC11021776 DOI: 10.3389/fmicb.2024.1326369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 03/12/2024] [Indexed: 04/19/2024] Open
Abstract
Leishmaniasis is a vector-borne disease caused by the protozoan parasite of Leishmania genus and is a complex disease affecting mostly tropical regions of the world. Unfortunately, despite the extensive effort made, there is no vaccine available for human use. Undoubtedly, a comprehensive understanding of the host-vector-parasite interaction is substantial for developing an effective prophylactic vaccine. Recently the role of sandfly saliva on disease progression has been uncovered which can make a substantial contribution in vaccine design. In this review we try to focus on the strategies that most probably meet the prerequisites of vaccine development (based on the current understandings) including live attenuated/non-pathogenic and subunit DNA vaccines. Innovative approaches such as reverse genetics, CRISP/R-Cas9 and antibiotic-free selection are now available to promisingly compensate for intrinsic drawbacks associated with these platforms. Our main goal is to call more attention toward the prerequisites of effective vaccine development while controlling the disease outspread is a substantial need.
Collapse
Affiliation(s)
- Negar Seyed
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | | | | |
Collapse
|
7
|
Nateghi-Rostami M, Sohrabi Y. Memory T cells: promising biomarkers for evaluating protection and vaccine efficacy against leishmaniasis. Front Immunol 2024; 15:1304696. [PMID: 38469319 PMCID: PMC10925770 DOI: 10.3389/fimmu.2024.1304696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/08/2024] [Indexed: 03/13/2024] Open
Abstract
Understanding the immune response to Leishmania infection and identifying biomarkers that correlate with protection are crucial for developing effective vaccines. One intriguing aspect of Leishmania infection is the persistence of parasites, even after apparent lesion healing. Various host cells, including dendritic cells, fibroblasts, and Langerhans cells, may serve as safe sites for latent infection. Memory T cells, especially tissue-resident memory T cells (TRM), play a crucial role in concomitant immunity against cutaneous Leishmania infections. These TRM cells are long-lasting and can protect against reinfection in the absence of persistent parasites. CD4+ TRM cells, in particular, have been implicated in protection against Leishmania infections. These cells are characterized by their ability to reside in the skin and rapidly respond to secondary infections by producing cytokines such as IFN-γ, which activates macrophages to kill parasites. The induction of CD4+ TRM cells has shown promise in experimental immunization, leading to protection against Leishmania challenge infections. Identifying biomarkers of protection is a critical step in vaccine development and CD4+ TRM cells hold potential as biomarkers, as their presence and functions may correlate with protection. While recent studies have shown that Leishmania-specific memory CD4+ T-cell subsets are present in individuals with a history of cutaneous leishmaniasis, further studies are needed to characterize CD4+ TRM cell populations. Overall, this review highlights the importance of memory T cells, particularly skin-resident CD4+ TRM cells, as promising targets for developing effective vaccines against leishmaniasis and as biomarkers of immune protection to assess the efficacy of candidate vaccines against human leishmaniasis.
Collapse
Affiliation(s)
| | - Yahya Sohrabi
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Westfälische Wilhelms-Universität, Münster, Germany
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czechia
| |
Collapse
|
8
|
Khandibharad S, Singh S. Synthetic biology for combating leishmaniasis. Front Microbiol 2024; 15:1338749. [PMID: 38362504 PMCID: PMC10867266 DOI: 10.3389/fmicb.2024.1338749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/08/2024] [Indexed: 02/17/2024] Open
Abstract
Leishmaniasis is a neglected tropical disease caused by protozoan parasites of the Leishmania genus. Despite the efforts to control and treat the disease, it still remains a major public health problem in many countries. Synthetic biology is a rapidly evolving interdisciplinary field that combines biology, engineering, and computer science to design and construct novel biological systems. In recent years, synthetic biology approaches have shown great promise for developing new and effective strategies to combat leishmaniasis. In this perspective, we summarize the recent advances in the use of synthetic biology for the development of vaccines, diagnostic tools, and novel therapeutics for leishmaniasis.
Collapse
Affiliation(s)
| | - Shailza Singh
- Systems Medicine Laboratory, National Centre for Cell Science, Pune, India
| |
Collapse
|
9
|
Kaye PM, Matlashewski G, Mohan S, Le Rutte E, Mondal D, Khamesipour A, Malvolti S. Vaccine value profile for leishmaniasis. Vaccine 2023; 41 Suppl 2:S153-S175. [PMID: 37951693 DOI: 10.1016/j.vaccine.2023.01.057] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/22/2022] [Accepted: 01/24/2023] [Indexed: 11/14/2023]
Abstract
Leishmania infections are global, occurring in 98 countries and all World Health Organization (WHO) regions with 600 million to 1 billion people at risk of infection. Visceral leishmaniasis is associated with almost 20,000 reported deaths annually, with children under 5 years of age being at the greatest risk of mortality. Amongst WHO-recognised Neglected Tropical Diseases (NTDs), leishmaniasis is one of the most important in terms of mortality and morbidity. With an increasing global burden of disease and a growing threat from climate change, urbanisation and drug resistance, there remains an imperative to develop leishmaniasis vaccines. New tools to understand correlates of protection and to assess vaccine efficacy are being developed to ease the transition into larger scale efficacy trials or provide alternate routes to licensure. Early indications suggest a diverse portfolio of manufacturers exists in endemic countries with an appetite to develop leishmaniasis vaccines. This Vaccine Value Profile (VVP) provides a high-level, comprehensive assessment of the currently available data to inform the potential public health, economic, and societal value of leishmaniasis vaccines. The leishmaniasis VVP was developed by a working group of subject matter experts from academia, public health groups, policy organizations, and non-profit organizations. All contributors have extensive expertise on various elements of the leishmaniasis VVP and have collectively described the state of knowledge and identified the current gaps. The VVP was developed using only existing and publicly available information.
Collapse
Affiliation(s)
- Paul M Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK.
| | - Greg Matlashewski
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada.
| | - Sakshi Mohan
- Center for Health Economics (CHE), University of York, York, UK.
| | - Epke Le Rutte
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland.
| | - Dinesh Mondal
- Laboratory Sciences and Services Division, International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh.
| | - Ali Khamesipour
- Center for Research and Training in Skin Diseases and Leprosy, Tehran University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
10
|
Oljuskin T, Azodi N, Volpedo G, Bhattacharya P, Markle HL, Hamano S, Matlashewski G, Satoskar AR, Gannavaram S, Nakhasi HL. Leishmania major centrin knock-out parasites reprogram tryptophan metabolism to induce a pro-inflammatory response. iScience 2023; 26:107593. [PMID: 37744403 PMCID: PMC10517402 DOI: 10.1016/j.isci.2023.107593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 07/07/2023] [Accepted: 08/07/2023] [Indexed: 09/26/2023] Open
Abstract
Leishmaniasis is a parasitic disease that is prevalent in 90 countries, and yet no licensed human vaccine exists against it. Toward control of leishmaniasis, we have developed Leishmania major centrin gene deletion mutant strains (LmCen-/-) as a live attenuated vaccine, which induces a strong IFN-γ-mediated protection to the host. However, the immune mechanisms of such protection remain to be understood. Metabolomic reprogramming of the host cells following Leishmania infection has been shown to play a critical role in pathogenicity and shaping the immune response following infection. Here, we applied untargeted mass spectrometric analysis to study the metabolic changes induced by infection with LmCen-/- and compared those with virulent L. major parasite infection to identify the immune mechanism of protection. Our data show that immunization with LmCen-/- parasites, in contrast to virulent L. major infection promotes a pro-inflammatory response by utilizing tryptophan to produce melatonin and downregulate anti-inflammatory kynurenine-AhR and FICZ-AhR signaling.
Collapse
Affiliation(s)
- Timur Oljuskin
- Animal Parasitic Diseases Laboratory, Agricultural Research Service, USDA, Beltsville, MD 20705, USA
| | - Nazli Azodi
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD 20993, USA
| | - Greta Volpedo
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Parna Bhattacharya
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD 20993, USA
| | - Hannah L. Markle
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD 20993, USA
| | - Shinjiro Hamano
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), The Joint Usage/Research Center on Tropical Disease, Nagasaki University, Nagasaki, Japan
- Nagasaki University Graduate School of Biomedical Sciences Doctoral Leadership Program, Nagasaki, Japan
| | - Greg Matlashewski
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Abhay R. Satoskar
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Sreenivas Gannavaram
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD 20993, USA
| | - Hira L. Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, CBER, FDA, Silver Spring, MD 20993, USA
| |
Collapse
|
11
|
Khandibharad S, Singh S. Immuno-metabolic signaling in leishmaniasis: insights gained from mathematical modeling. BIOINFORMATICS ADVANCES 2023; 3:vbad125. [PMID: 37799190 PMCID: PMC10548086 DOI: 10.1093/bioadv/vbad125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/27/2023] [Accepted: 09/12/2023] [Indexed: 10/07/2023]
Abstract
Motivation Leishmaniasis is a global concern especially in underdeveloped and developing subtropical and tropical regions. The extent of infectivity in host is majorly dependent on functional polarization of macrophages. Classically activated M1 macrophage can eliminate parasite through production of iNOS and alternatively activated M2 macrophages can promote parasite growth through by providing shelter and nutrients to parasite. The biological processes involved in immune signaling and metabolism of host and parasite might be responsible for deciding fate of parasite. Results Using systems biology approach, we constructed two mathematical models and inter-regulatory immune-metabolic networks of M1 and M2 state, through which we identified crucial components that are associated with these phenotypes. We also demonstrated how parasite may modulate M1 phenotype for its growth and proliferation and transition to M2 state. Through our previous findings as well as from recent findings we could identify SHP-1 as a key component in regulating the immune-metabolic characterization of M2 macrophage. By targeting SHP-1 at cellular level, it might be possible to modulate immuno-metabolic mechanism and thereby control parasite survival. Availability and implementation Mathematical modeling is implemented as a workflow and the models are deposited in BioModel database. FactoMineR is available at: https://github.com/cran/FactoMineR/tree/master.
Collapse
Affiliation(s)
- Shweta Khandibharad
- Systems Medicine Laboratory, National Centre for Cell Science, NCCS Complex, SPPU Campus, Pune 411007, India
| | - Shailza Singh
- Systems Medicine Laboratory, National Centre for Cell Science, NCCS Complex, SPPU Campus, Pune 411007, India
| |
Collapse
|
12
|
Costa CHN, Chang KP, Costa DL, Cunha FVM. From Infection to Death: An Overview of the Pathogenesis of Visceral Leishmaniasis. Pathogens 2023; 12:969. [PMID: 37513817 PMCID: PMC10384967 DOI: 10.3390/pathogens12070969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/02/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Kala-azar, also known as visceral leishmaniasis (VL), is a disease caused by Leishmania infantum and L. donovani. Patients experience symptoms such as fever, weight loss, paleness, and enlarged liver and spleen. The disease also affects immunosuppressed individuals and has an overall mortality rate of up to 10%. This overview explores the literature on the pathogenesis of preclinical and clinical stages, including studies in vitro and in animal models, as well as complications and death. Asymptomatic infection can result in long-lasting immunity. VL develops in a minority of infected individuals when parasites overcome host defenses and multiply in tissues such as the spleen, liver, and bone marrow. Hepatosplenomegaly occurs due to hyperplasia, resulting from parasite proliferation. A systemic inflammation mediated by cytokines develops, triggering acute phase reactants from the liver. These cytokines can reach the brain, causing fever, cachexia and vomiting. Similar to sepsis, disseminated intravascular coagulation (DIC) occurs due to tissue factor overexpression. Anemia, hypergammaglobulinemia, and edema result from the acute phase response. A regulatory response and lymphocyte depletion increase the risk of bacterial superinfections, which, combined with DIC, are thought to cause death. Our understanding of VL's pathogenesis is limited, and further research is needed to elucidate the preclinical events and clinical manifestations in humans.
Collapse
Affiliation(s)
- Carlos H N Costa
- Centro de Investigações em Agravos Tropicais Emergentes e Negligenciados, Instituto de Doenças Tropicais Natan Portella, Universidade Federal do Piauí, Rua Artur de Vasconcelos 151-Sul, Teresina 64002-510, PI, Brazil
| | - Kwang-Poo Chang
- Department of Microbiology/Immunology, Center for Cancer Cell Biology, Immunology & Infection, Chicago Medical School, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Dorcas L Costa
- Centro de Investigações em Agravos Tropicais Emergentes e Negligenciados, Instituto de Doenças Tropicais Natan Portella, Universidade Federal do Piauí, Rua Artur de Vasconcelos 151-Sul, Teresina 64002-510, PI, Brazil
| | - Francisco Valmor M Cunha
- Departament of Physiotherapy, Centro Universitário Uninovafapi, Rua Vitorino Orthiges Fernandes, 6123-Uruguai, Teresina 64073-505, PI, Brazil
| |
Collapse
|
13
|
Moreira POL, Nogueira PM, Monte-Neto RL. Next-Generation Leishmanization: Revisiting Molecular Targets for Selecting Genetically Engineered Live-Attenuated Leishmania. Microorganisms 2023; 11:microorganisms11041043. [PMID: 37110466 PMCID: PMC10145799 DOI: 10.3390/microorganisms11041043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/07/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Despite decades of research devoted to finding a vaccine against leishmaniasis, we are still lacking a safe and effective vaccine for humans. Given this scenario, the search for a new prophylaxis alternative for controlling leishmaniasis should be a global priority. Inspired by leishmanization-a first generation vaccine strategy where live L. major parasites are inoculated in the skin to protect against reinfection-live-attenuated Leishmania vaccine candidates are promising alternatives due to their robust elicited protective immune response. In addition, they do not cause disease and could provide long-term protection upon challenge with a virulent strain. The discovery of a precise and easy way to perform CRISPR/Cas-based gene editing allowed the selection of safer null mutant live-attenuated Leishmania parasites obtained by gene disruption. Here, we revisited molecular targets associated with the selection of live-attenuated vaccinal strains, discussing their function, their limiting factors and the ideal candidate for the next generation of genetically engineered live-attenuated Leishmania vaccines to control leishmaniasis.
Collapse
Affiliation(s)
- Paulo O L Moreira
- Biotechnology Applied to Pathogens (BAP), Instituto René Rachou, Fundação Oswaldo Cruz, Fiocruz Minas, Belo Horizonte 30190-009, Brazil
| | - Paula M Nogueira
- Biotechnology Applied to Pathogens (BAP), Instituto René Rachou, Fundação Oswaldo Cruz, Fiocruz Minas, Belo Horizonte 30190-009, Brazil
| | - Rubens L Monte-Neto
- Biotechnology Applied to Pathogens (BAP), Instituto René Rachou, Fundação Oswaldo Cruz, Fiocruz Minas, Belo Horizonte 30190-009, Brazil
| |
Collapse
|
14
|
Vats K, Tandon R, Roshanara, Beg MA, Corrales RM, Yagoubat A, Reyaz E, Wani TH, Baig MS, Chaudhury A, Krishnan A, Puri N, Salotra P, Sterkers Y, Selvapandiyan A. Interaction of novel proteins, centrin4 and protein of centriole in Leishmania parasite and their effects on the parasite growth. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119416. [PMID: 36623775 DOI: 10.1016/j.bbamcr.2022.119416] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 12/07/2022] [Accepted: 12/14/2022] [Indexed: 01/08/2023]
Abstract
Centrins are cytoskeletal proteins associated with the centrosomes or basal bodies in the eukaryotes. We previously reported the involvement of Centrin 1-3 proteins in cell division in the protozoan parasites Leishmania donovani and Trypanosoma brucei. Centrin4 and 5, unique to such parasites, had never been characterized in Leishmania parasite. In the current study, we addressed the function of centrin4 (LdCen4) in Leishmania. By dominant-negative study, the episomal expression of C-terminal truncated LdCen4 in the parasite reduced the parasite growth. LdCen4 double allele gene deletion by either homologous recombination or CRISPR-Cas9 was not successful in L. donovani. However, CRISPR-Cas9-based deletion of the homologous gene was possible in L. mexicana, which attenuated the parasite growth in vitro, but not ex vivo in the macrophages. LdCen4 also interacts with endogenous and overexpressed LdPOC protein, a homolog of centrin reacting human POC (protein of centriole) in a calcium sensitive manner. LdCen4 and LdPOC binding has also been confirmed through in silico analysis by protein structural docking and validated by co-immunoprecipitation. By immunofluorescence studies, we found that both the proteins share a common localization at the basal bodies. Thus, for the first time, this article describes novel centrin4 and its binding protein in the protozoan parasites.
Collapse
Affiliation(s)
- Kavita Vats
- Department of Molecular Medicine, Jamia Hamdard, New Delhi 110062, India; Department of Bio & Nano Technology, Bio & Nano Technology Centre, Guru Jambheshwar University of Science and Technology, Hisar 125001, India; MiVEGEC, University of Montpellier, CNRS, IRD, Academic Hospital (CHU) of Montpellier, Montpellier 34295, France
| | - Rati Tandon
- Department of Molecular Medicine, Jamia Hamdard, New Delhi 110062, India
| | - Roshanara
- Department of Molecular Medicine, Jamia Hamdard, New Delhi 110062, India
| | - Mirza A Beg
- Department of Molecular Medicine, Jamia Hamdard, New Delhi 110062, India
| | - Rosa M Corrales
- MiVEGEC, University of Montpellier, CNRS, IRD, Academic Hospital (CHU) of Montpellier, Montpellier 34295, France
| | - Akila Yagoubat
- MiVEGEC, University of Montpellier, CNRS, IRD, Academic Hospital (CHU) of Montpellier, Montpellier 34295, France
| | - Enam Reyaz
- Department of Molecular Medicine, Jamia Hamdard, New Delhi 110062, India
| | - Tasaduq H Wani
- Department of Molecular Medicine, Jamia Hamdard, New Delhi 110062, India
| | - Mirza S Baig
- Department of Molecular Medicine, Jamia Hamdard, New Delhi 110062, India
| | - Ashok Chaudhury
- Department of Bio & Nano Technology, Bio & Nano Technology Centre, Guru Jambheshwar University of Science and Technology, Hisar 125001, India
| | - Anuja Krishnan
- Department of Molecular Medicine, Jamia Hamdard, New Delhi 110062, India
| | - Niti Puri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Poonam Salotra
- ICMR-National Institute of Pathology, Safdarjung Hospital Campus, New Delhi 110029, India
| | - Yvon Sterkers
- MiVEGEC, University of Montpellier, CNRS, IRD, Academic Hospital (CHU) of Montpellier, Montpellier 34295, France
| | | |
Collapse
|
15
|
Bandi C, Mendoza-Roldan JA, Otranto D, Alvaro A, Louzada-Flores VN, Pajoro M, Varotto-Boccazzi I, Brilli M, Manenti A, Montomoli E, Zuccotti G, Epis S. Leishmania tarentolae: a vaccine platform to target dendritic cells and a surrogate pathogen for next generation vaccine research in leishmaniases and viral infections. Parasit Vectors 2023; 16:35. [PMID: 36703216 PMCID: PMC9879565 DOI: 10.1186/s13071-023-05651-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/03/2023] [Indexed: 01/27/2023] Open
Abstract
Parasites of the genus Leishmania are unusual unicellular microorganisms in that they are characterized by the capability to subvert in their favor the immune response of mammalian phagocytes, including dendritic cells. Thus, in overt leishmaniasis, dendritic cells and macrophages are converted into a niche for Leishmania spp. in which the parasite, rather than being inactivated and disassembled, survives and replicates. In addition, Leishmania parasites hitchhike onto phagocytic cells, exploiting them as a mode of transport to lymphoid tissues where other phagocytic cells are potentially amenable to parasite colonization. This propensity of Leishmania spp. to target dendritic cells has led some researchers to consider the possibility that the non-pathogenic, reptile-associated Leishmania tarentolae could be exploited as a vaccine platform and vehicle for the production of antigens from different viruses and for the delivery of the antigens to dendritic cells and lymph nodes. In addition, as L. tarentolae can also be regarded as a surrogate of pathogenic Leishmania parasites, this parasite of reptiles could possibly be developed into a vaccine against human and canine leishmaniases, exploiting its immunological cross-reactivity with other Leishmania species, or, after its engineering, for the expression of antigens from pathogenic species. In this article we review published studies on the use of L. tarentolae as a vaccine platform and vehicle, mainly in the areas of leishmaniases and viral infections. In addition, a short summary of available knowledge on the biology of L. tarentolae is presented, together with information on the use of this microorganism as a micro-factory to produce antigens suitable for the serodiagnosis of viral and parasitic infections.
Collapse
Affiliation(s)
- Claudio Bandi
- grid.4708.b0000 0004 1757 2822Department of Biosciences, Pediatric CRC “Romeo ed Enrica Invernizzi”–University of Milan, Milan, Italy
| | | | - Domenico Otranto
- grid.7644.10000 0001 0120 3326Department of Veterinary Medicine, University of Bari, Valenzano, Italy
| | - Alessandro Alvaro
- grid.4708.b0000 0004 1757 2822Department of Biosciences, Pediatric CRC “Romeo ed Enrica Invernizzi”–University of Milan, Milan, Italy
| | | | - Massimo Pajoro
- grid.4708.b0000 0004 1757 2822Department of Biosciences, Pediatric CRC “Romeo ed Enrica Invernizzi”–University of Milan, Milan, Italy
| | - Ilaria Varotto-Boccazzi
- grid.4708.b0000 0004 1757 2822Department of Biosciences, Pediatric CRC “Romeo ed Enrica Invernizzi”–University of Milan, Milan, Italy
| | - Matteo Brilli
- grid.4708.b0000 0004 1757 2822Department of Biosciences, Pediatric CRC “Romeo ed Enrica Invernizzi”–University of Milan, Milan, Italy
| | | | - Emanuele Montomoli
- grid.511037.1VisMederi, Siena, Italy ,grid.9024.f0000 0004 1757 4641Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Gianvincenzo Zuccotti
- grid.4708.b0000 0004 1757 2822Department of Biomedical and Clinical Sciences, Pediatric CRC “Romeo ed Enrica Invernizzi”–University of Milan, Milan, Italy ,Department of Pediatrics, Ospedale dei Bambini-Buzzi, Milan, Italy
| | - Sara Epis
- grid.4708.b0000 0004 1757 2822Department of Biosciences, Pediatric CRC “Romeo ed Enrica Invernizzi”–University of Milan, Milan, Italy
| |
Collapse
|
16
|
Duthie MS, Machado BAS, Badaró R, Kaye PM, Reed SG. Leishmaniasis Vaccines: Applications of RNA Technology and Targeted Clinical Trial Designs. Pathogens 2022; 11:pathogens11111259. [PMID: 36365010 PMCID: PMC9695603 DOI: 10.3390/pathogens11111259] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/20/2022] [Accepted: 10/27/2022] [Indexed: 11/17/2022] Open
Abstract
Leishmania parasites cause a variety of discrete clinical diseases that present in regions where their specific sand fly vectors sustain transmission. Clinical and laboratory research indicate the potential of immunization to prevent leishmaniasis and a wide array of vaccine candidates have been proposed. Unfortunately, multiple factors have precluded advancement of more than a few Leishmania targeting vaccines to clinical trial. The recent maturation of RNA vaccines into licensed products in the context of COVID-19 indicates the likelihood of broader use of the technology. Herein, we discuss the potential benefits provided by RNA technology as an approach to address the bottlenecks encountered for Leishmania vaccines. Further, we outline a variety of strategies that could be used to more efficiently evaluate Leishmania vaccine efficacy, including controlled human infection models and initial use in a therapeutic setting, that could prioritize candidates before evaluation in larger, longer and more complicated field trials.
Collapse
Affiliation(s)
| | - Bruna A S Machado
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, Bahia, Brazil
| | - Roberto Badaró
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, Bahia, Brazil
| | - Paul M Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York YO10 5DD, UK
| | - Steven G Reed
- HDT Bio, 1616 Eastlake Ave E, Seattle, WA 98102, USA
| |
Collapse
|
17
|
Kumari D, Mahajan S, Kour P, Singh K. Virulence factors of Leishmania parasite: Their paramount importance in unraveling novel vaccine candidates and therapeutic targets. Life Sci 2022; 306:120829. [PMID: 35872004 DOI: 10.1016/j.lfs.2022.120829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/15/2022] [Accepted: 07/17/2022] [Indexed: 12/30/2022]
Abstract
Leishmaniasis is a neglected tropical disease and remains a global concern for healthcare. It is caused by an opportunistic protozoan parasite belonging to the genus Leishmania and affects millions worldwide. This disease is mainly prevalent in tropical and subtropical regions and is associated with a high risk of public morbidity and mortality if left untreated. Transmission of this deadly disease is aggravated by the bite of female sand-fly vectors (Phlebotomus and Lutzomyia). With time, significant advancement in leishmaniasis-related research has been carried out to cope with the disease burden. Still, the Leishmania parasite has also co-evolved with its host and adapted successfully within the host's lethal milieu/environment. Thus, understanding and knowledge of various leishmanial virulence factors responsible for the parasitic infection are essential for exploring drug targets and vaccine candidates. The present review elucidates the importance of virulence factors in pathogenesis and summarizes the major leishmanial virulence molecules contributing to the parasitic infection during host-pathogen interaction. Furthermore, we have also elaborated on the potential contribution of leishmanial virulence proteins in developing vaccine candidates and exploring novel therapeutics against this parasitic disease. We aim to represent a clearer picture of parasite pathogenesis within the human host that can further aid in unraveling new strategies to fight against the deadly infection of leishmaniasis.
Collapse
Affiliation(s)
- Diksha Kumari
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shavi Mahajan
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Parampreet Kour
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Kuljit Singh
- Infectious Diseases Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|