1
|
Barrie U, Floyd K, Datta A, Wetzel DM. MAPK/ERK activation in macrophages promotes Leishmania internalization and pathogenesis. Microbes Infect 2024; 26:105353. [PMID: 38763478 DOI: 10.1016/j.micinf.2024.105353] [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] [Academic Contribution Register] [Received: 01/18/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/21/2024]
Abstract
The obligate intracellular parasite Leishmania binds several receptors to trigger uptake by phagocytic cells, ultimately resulting in visceral or cutaneous leishmaniasis. A series of signaling pathways in host cells, which are critical for establishment and persistence of infection, are activated during Leishmania internalization. Thus, preventing Leishmania uptake by phagocytes could be a novel therapeutic strategy for leishmaniasis. However, the host cellular machinery mediating promastigote and amastigote uptake is not well understood. Here, using small molecule inhibitors of Mitogen-activated protein/Extracellular signal regulated kinases (MAPK/ERK), we demonstrate that ERK1/2 mediates Leishmania amazonensis uptake and (to a lesser extent) phagocytosis of beads by macrophages. We find that inhibiting host MEK1/2 or ERK1/2 leads to inefficient amastigote uptake. Moreover, using inhibitors and primary macrophages lacking spleen tyrosine kinase (SYK) or Abl family kinases, we show that SYK and Abl family kinases mediate Raf, MEK, and ERK1/2 activity and are necessary for uptake. Finally, we demonstrate that trametinib, a MEK1/2 inhibitor used to treat cancer, reduces disease severity and parasite burden in Leishmania-infected mice, even if it is started after lesions develop. Our results show that maximal Leishmania infection requires MAPK/ERK and highlight potential for MAPK/ERK-mediated signaling pathways to be novel therapeutic targets for leishmaniasis.
Collapse
Affiliation(s)
- Umaru Barrie
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, United States; Medical Scientist Training Program, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, United States
| | - Katherine Floyd
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, United States
| | - Arani Datta
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, United States
| | - Dawn M Wetzel
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, United States; Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, United States.
| |
Collapse
|
2
|
Reyaz E, Puri N, Selvapandiyan A. Global Remodeling of Host Proteome in Response to Leishmania Infection. ACS Infect Dis 2024; 10:5-19. [PMID: 38084821 DOI: 10.1021/acsinfecdis.3c00492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/13/2024]
Abstract
The protozoan parasite Leishmania possesses an intrinsic ability to modulate a multitude of pathways in the host, toward aiding its own proliferation. In response, the host reprograms its cellular, immunological, and metabolic machinery to evade the parasite's lethal impact. Besides inducing various antioxidant signaling pathways to counter the elevated stress response proteins like heme oxygenase-1 (HO-1), Leishmania also attempts to delay host cell apoptosis by promoting anti-apoptotic proteins like Bcl-2. The downstream modulation of apoptotic proteins is regulated by effector pathways, including the PI3K/Akt survival pathway, the mitogen-activated protein kinases (MAPKs) signaling pathway, and STAT phosphorylation. In addition, Leishmania assists in its infection in a time-dependent manner by modulating the level of various proteins of autophagic machinery. Immune effector cells, such as mast cells and neutrophils, entrap and kill the pathogen by secreting various granular proteins. In contrast, the host macrophages exert their leishmanicidal effect by secreting various cytokines, such as IL-2, IL-12, etc. An interplay of various signaling pathways occurs in an organized network that is highly specific to both pathogen and host species. This Review analyzes the modulation of expression of proteins, including the cytokines, providing a realistic approach toward understanding the pathophysiology of disease and predicting some prominent markers for disease intervention and vaccine support strategies.
Collapse
Affiliation(s)
- Enam Reyaz
- Department of Molecular Medicine, Jamia Hamdard, New Delhi 110062, India
| | - Niti Puri
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | | |
Collapse
|
3
|
Milano M, Agapito G, Cannataro M. An Exploratory Application of Multilayer Networks and Pathway Analysis in Pharmacogenomics. Genes (Basel) 2023; 14:1915. [PMID: 37895264 PMCID: PMC10606656 DOI: 10.3390/genes14101915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/04/2023] [Revised: 09/26/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Over the years, network analysis has become a promising strategy for analysing complex system, i.e., systems composed of a large number of interacting elements. In particular, multilayer networks have emerged as a powerful framework for modelling and analysing complex systems with multiple types of interactions. Network analysis can be applied to pharmacogenomics to gain insights into the interactions between genes, drugs, and diseases. By integrating network analysis techniques with pharmacogenomic data, the goal consists of uncovering complex relationships and identifying key genes to use in pathway enrichment analysis to figure out biological pathways involved in drug response and adverse reactions. In this study, we modelled omics, disease, and drug data together through multilayer network representation. Then, we mined the multilayer network with a community detection algorithm to obtain the top communities. After that, we used the identified list of genes from the communities to perform pathway enrichment analysis (PEA) to figure out the biological function affected by the selected genes. The results show that the genes forming the top community have multiple roles through different pathways.
Collapse
Affiliation(s)
- Marianna Milano
- Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy
- Data Analytics Research Center, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy; (G.A.); (M.C.)
| | - Giuseppe Agapito
- Data Analytics Research Center, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy; (G.A.); (M.C.)
- Department of Law, Economics and Social Sciences, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy
| | - Mario Cannataro
- Data Analytics Research Center, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy; (G.A.); (M.C.)
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
4
|
Chen H, Liu L, Li M, Zhu D, Tian G. Epicardial Adipose Tissue-Derived Leptin Promotes Myocardial Injury in Metabolic Syndrome Rats Through PKC/NADPH Oxidase/ROS Pathway. J Am Heart Assoc 2023; 12:e029415. [PMID: 37489731 PMCID: PMC10492984 DOI: 10.1161/jaha.123.029415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/07/2023] [Accepted: 06/19/2023] [Indexed: 07/26/2023]
Abstract
Background The epicardial adipose tissue (EAT) of metabolic syndrome (MetS) is abnormally accumulated with dysfunctional secretion of adipokines, closely relating to cardiac dysfunction. The current study was designed to identify the effects of EAT-derived leptin on the myocardium of MetS rats and explore the potential molecular mechanisms. Methods and Results A MetS rat model was established in 8-week-old Wistar rats by a 12-week high-fat diet. MetS rats exhibited increased leptin secretion from EAT, cardiac hypertrophy, and diastolic dysfunction with preserved systolic function. The myocardium of MetS rats had abnormal structure, increased oxidative stress injury, and higher inflammatory factor levels, especially the subepicardial myocardium, which was correlated with the EAT-derived leptin level but not the serum leptin. The EAT was separated from each group of rats to prepare EAT-conditioned medium. H9C2 rat cardiomyoblasts were treated with EAT-conditioned medium or leptin, plus various inhibitors. EAT-derived leptin from MetS rats promoted mitochondrial oxidative stress and dysfunction, induced mitochondrial pathway apoptosis, and inhibited cell viability in H9C2 cardiomyoblasts via the protein kinase C/reduced nicotinamide adenine dinucleotide phosphate oxidase/reactive oxygen species (PKC/NADPH oxidase/ROS) pathway. EAT-derived leptin from MetS rats stimulated inflammation in H9C2 cardiomyocytes by promoting activator protein 1 nuclear translocation via the PKC/NADPH oxidase/ROS pathway. Leptin promoted the interaction between p-p47phox and gp91phox in H9C2 cardiomyocytes via protein kinase C, activating nicotinamide adenine dinucleotide phosphate oxidase, increasing reactive oxygen species generation, and inhibiting cell viability. Conclusions EAT-derived leptin induces MetS-related myocardial injury through the following 2 cooperative ways via PKC/NADPH oxidase/ROS pathway: (1) inducing mitochondrial pathway apoptosis by promoting mitochondrial oxidative stress and dysfunction; and (2) stimulating inflammation by promoting activator protein 1 nuclear translocation.
Collapse
Affiliation(s)
- Hui Chen
- Heart Center of Henan Provincial People’s Hospital, Central China Fuwai HospitalCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Lei Liu
- Department of CardiologyThe First Affiliated Hospital of Xi’an Jiao Tong UniversityXi’anShaanxiChina
| | - Min Li
- Department of CardiologyThe First Affiliated Hospital of Xi’an Jiao Tong UniversityXi’anShaanxiChina
| | - Danjun Zhu
- Department of CardiologyThe First Affiliated Hospital of Xi’an Jiao Tong UniversityXi’anShaanxiChina
| | - Gang Tian
- Department of CardiologyThe First Affiliated Hospital of Xi’an Jiao Tong UniversityXi’anShaanxiChina
| |
Collapse
|
5
|
Gupta D, Singh PK, Yadav PK, Narender T, Patil UK, Jain SK, Chourasia MK. Emerging strategies and challenges of molecular therapeutics in antileishmanial drug development. Int Immunopharmacol 2023; 115:109649. [PMID: 36603357 DOI: 10.1016/j.intimp.2022.109649] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/29/2022] [Revised: 12/16/2022] [Accepted: 12/24/2022] [Indexed: 01/05/2023]
Abstract
Molecular therapy refers to targeted therapies based on molecules which have been intelligently directed towards specific biomolecular structures and include small molecule drugs, monoclonal antibodies, proteins and peptides, DNA or RNA-based strategies, targeted chemotherapy and nanomedicines. Molecular therapy is emerging as the most effective strategy to combat the present challenges of life-threatening visceral leishmaniasis, where the successful human vaccine is currently unavailable. Moreover, current chemotherapy-based strategies are associated with the issues of ineffective targeting, unavoidable toxicities, invasive therapies, prolonged treatment, high treatment costs and the development of drug-resistant strains. Thus, the rational approach to antileishmanial drug development primarily demands critical exploration and exploitation of biochemical differences between host and parasite biology, immunocharacteristics of parasite homing, and host-parasite interactions at the molecular/cellular level. Following this, the novel technology-based designing and development of host and/or parasite-targeted therapeutics having leishmanicidal and immunomodulatory activity is utmost essential to improve treatment efficacy. Thus, the present review is focused on immunological and molecular checkpoint targets in host-pathogen interaction, and molecular therapeutic prospects for Leishmania intervention, and the challenges ahead.
Collapse
Affiliation(s)
- Deepak Gupta
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar 470003, M.P., India; Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Pankaj K Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, Telangana, India
| | - Pavan K Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Tadigoppula Narender
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Umesh K Patil
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar 470003, M.P., India
| | - Sanjay K Jain
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar 470003, M.P., India
| | - Manish K Chourasia
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India.
| |
Collapse
|
6
|
Khandibharad S, Singh S. Artificial intelligence channelizing protein-peptide interactions pipeline for host-parasite paradigm in IL-10 and IL-12 reciprocity by SHP-1. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166466. [PMID: 35750267 DOI: 10.1016/j.bbadis.2022.166466] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/05/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022]
Abstract
Identification of molecular targets in any cellular phenomena is a challenge and a path that one endeavors upon independently. We have identified a phosphatase SHP-1 as a point of intervention of IL-10 and IL-12 reciprocity in leishmaniasis. The therapeutic model that we have developed uniquely targets this protein but the pipeline in general can be used by the researchers for their unique targets. Naturally occurring peptides are well known for their biochemical participation in cellular functions hence we were motivated to use this uniqueness of physico-chemical properties of peptides conferred by amino acids through machine learning to channelize a mode of therapeutic exploration in infectious disease. Using computational approaches, we identified high order sequence conservation and similarity in SHP-1 sequence which was also evolutionarily conserved, complete structure of Mouse SHP-1 was predicted and validated, a unique motif of the same was identified against which library of synthetic peptides were designed and validated followed by screening the library by docking them with MuSHP-1 protein structure. Our findings showed 3 peptides had high binding affinity and in future can be validated using cell based and in vivo assays.
Collapse
Affiliation(s)
- Shweta Khandibharad
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, INDIA
| | - Shailza Singh
- National Centre for Cell Science, NCCS Complex, Ganeshkhind, SP Pune University Campus, Pune 411007, INDIA.
| |
Collapse
|
7
|
Liu Y, Dai M, Yang P, Cao L, Lu L. Src-homology domain 2 containing protein tyrosine phosphatase-1 (SHP-1) directly binds to proto-oncogene tyrosine-protein kinase Src (c-Src) and promotes the transcriptional activation of connexin 43 (Cx43). Bioengineered 2022; 13:13534-13543. [PMID: 35659197 PMCID: PMC9276044 DOI: 10.1080/21655979.2022.2079252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/09/2022] Open
Abstract
The prevalence of atrial fibrillation (AF), which is one of the common arrhythmias in clinics, is increasing sharply and has affected millions of patients, which is expected to triple by 2050. The purpose of the study was to explore the regulatory relationship between Src-homology domain 2 containing protein tyrosine phosphatase-1 (SHP-1) and proto-oncogene tyrosine-protein kinase Src (c-Src) and the regulation of Connexins 43 (Cx43), and its effect on AF was also studied. Mouse atrial myocyte line (HL-1 cell line) was used as the research object. After overexpression of SHP-1, the expressions of p-c-Src, Cx43, and SHP-1 were detected by Western blot and cellular immunofluorescence, respectively. The location and interaction of SHP-1 and c-Src in the cells were detected by immunofluorescence co-localization and co-immunoprecipitation (Co-IP). The regulation of c-Src and Cx43 was detected by DNA pull down, chromatin co-immunoprecipitation (CHIP), and dual-luciferase reporter system. The results revealed that overexpression of SHP-1 could inhibit the phosphorylation and activation of c-Src and increase the expression of Cx43. Moreover, there was a direct binding between SHP-1 and c-Src, and c-Src could bind to the promoter region of Cx43 and inhibit the transcription of Cx43. In conclusion, SHP-1 could bind to c-Src and inhibit the activity of c-Src, thus enhancing the transcriptional activation of Cx43 and improving the function of gap junction.
Collapse
Affiliation(s)
- YiHao Liu
- Department of Cardiovascular Medicine, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Meng Dai
- Department of Palliative Medicine, Chongqing University Cancer Hospital, Chongqing, China
| | - PengHui Yang
- Department of Cardiovascular Medicine, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Li Cao
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Lu
- Department of Critical Care Medicine, University-Town Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
8
|
Gupta AK, Das S, Kamran M, Ejazi SA, Ali N. The Pathogenicity and Virulence of Leishmania - interplay of virulence factors with host defenses. Virulence 2022; 13:903-935. [PMID: 35531875 PMCID: PMC9154802 DOI: 10.1080/21505594.2022.2074130] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/28/2022] Open
Abstract
Leishmaniasis is a group of disease caused by the intracellular protozoan parasite of the genus Leishmania. Infection by different species of Leishmania results in various host immune responses, which usually lead to parasite clearance and may also contribute to pathogenesis and, hence, increasing the complexity of the disease. Interestingly, the parasite tends to reside within the unfriendly environment of the macrophages and has evolved various survival strategies to evade or modulate host immune defense. This can be attributed to the array of virulence factors of the vicious parasite, which target important host functioning and machineries. This review encompasses a holistic overview of leishmanial virulence factors, their role in assisting parasite-mediated evasion of host defense weaponries, and modulating epigenetic landscapes of host immune regulatory genes. Furthermore, the review also discusses the diagnostic potential of various leishmanial virulence factors and the advent of immunomodulators as futuristic antileishmanial drug therapy.
Collapse
Affiliation(s)
- Anand Kumar Gupta
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Sonali Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Mohd Kamran
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Sarfaraz Ahmad Ejazi
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Nahid Ali
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| |
Collapse
|
9
|
Ndzeidze R, Leestemaker-Palmer A, Danelishvili L, Bermudez LE. Virulent Mycobacterium avium subspecies hominissuis subverts macrophages during early stages of infection. MICROBIOLOGY (READING, ENGLAND) 2022; 168. [PMID: 35133955 DOI: 10.1099/mic.0.001133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 11/18/2022]
Abstract
Virulent non-tuberculous Mycobacteria (NTMs) successfully reside and multiply within the phagosomes of phagocytic cells such as monocytes and macrophages. Macrophages play a very important role in the innate clearance of intracellular pathogens including NTMs. Attenuated Mycobacterium avium subsp. hominissuis 100 enters macrophages but is incapable of escaping these cells via canonical mycobacteria escape mechanisms. Alternatively, virulent Mycobacterium avium subsp. hominissuis 104 and Mycobacterium abscessus subsp. abscessus are able to modify macrophages to suit their growth, survival and ultimately escape from macrophages, while non-virulent Mycobacterium smegmatis is readily killed by macrophages. In this study we focused on early infection of macrophages with NTMs to determine the phenotypic response of macrophages, M1 or M2 differentiation, and phosphorylation alterations that can affect cellular response to invading bacteria. Our findings indicate that infection of the macrophage with MAH 100 and M. smegmatis favours the development of M1 macrophage, a pro-inflammatory phenotype associated with the killing of intracellular pathogens, while infection of the macrophage with MAH 104 and M. abscessus favoured the development of M2 macrophage, an anti-inflammatory phenotype associated with the healing process. Interference with the host post-translational mechanisms, such as protein phosphorylation, is a key strategy used by many intracellular bacterial pathogens to modulate macrophage phenotype and subvert macrophage function. By comparing protein phosphorylation patterns of infected macrophages, we observed that uptake of both MAH 100 and M. smegmatis resulted in MARCKS-related protein phosphorylation, which has been associated with macrophage activation. In contrast, in macrophages infected with MAH 104 and M. abscessus, methionine adenosyltransferase IIβ, an enzyme that catalyses the biosynthesis of S-adenosylmethionine, a methyl donor for DNA methylation. Inhibition of DNA methylation with 5-aza-2 deoxycytidine, significantly impaired the survival of MAH 104 in macrophages. Our findings suggest that the virulent MAH 104 and M. abscessus enhance its survival in the macrophage possibly through interference with the epigenome responses.
Collapse
Affiliation(s)
- Robert Ndzeidze
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Corvallis, OR, USA
| | - Amy Leestemaker-Palmer
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Corvallis, OR, USA
| | - Lia Danelishvili
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Corvallis, OR, USA
| | - Luiz E Bermudez
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Corvallis, OR, USA.,Department of Microbiology, College of Sciences, Oregon State University, Corvallis, OR, USA
| |
Collapse
|
10
|
Khandibharad S, Singh S. Computational System Level Approaches for Discerning Reciprocal Regulation of IL10 and IL12 in Leishmaniasis. Front Genet 2022; 12:784664. [PMID: 35126456 PMCID: PMC8807686 DOI: 10.3389/fgene.2021.784664] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/28/2021] [Accepted: 12/20/2021] [Indexed: 12/22/2022] Open
Abstract
IL12 and IL10 are two of the major cytokines which control the fate of Leishmaniasis. This paper presents two models healthy state and diseased state which shows how secretion of IL12 is responsible for parasite elimination and IL10 can jeopardize the parasite elimination and promote its survival. Epigenetic modification in the host IL12 and IL10 promoter can decide the fate of parasites. It was observed that reciprocal relationship exists between IL12 and IL10 and that is majorly controlled by a transcription factor NFAT5 from Rel family of transcription factors. By targeting this transcription factor at the cellular level, it might be possible to modulate the release of powerful pro-inflammatory cytokines, thereby reducing parasite survival. The mathematical models developed here serves as a step towards finding a key component that can pave a way for therapeutic investigation.
Collapse
|
11
|
Sanz CR, Miró G, Sevane N, Reyes-Palomares A, Dunner S. Modulation of Host Immune Response during Leishmania infantum Natural Infection: A Whole-Transcriptome Analysis of the Popliteal Lymph Nodes in Dogs. Front Immunol 2022; 12:794627. [PMID: 35058931 PMCID: PMC8763708 DOI: 10.3389/fimmu.2021.794627] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/13/2021] [Accepted: 12/10/2021] [Indexed: 12/21/2022] Open
Abstract
Leishmania infantum, the etiological agent of canine leishmaniosis (CanL) in Europe, was responsible of the largest outbreak of human leishmaniosis in Spain. The parasite infects and survives within myeloid lineage cells, causing a potentially fatal disease if left untreated. The only treatment option relies on chemotherapy, although immunotherapy strategies are being considered as novel approaches to prevent progression of the disease. To this aim, a deeper characterization of the molecular mechanisms behind the immunopathogenesis of leishmaniosis is necessary. Thus, we evaluated, for the first time, the host immune response during L. infantum infection through transcriptome sequencing of the popliteal lymph nodes aspirates of dogs with CanL. Differential expression and weighted gene co-expression network analyses were performed, resulting in the identification of 5,461 differentially expressed genes (DEGs) and four key modules in sick dogs, compared to controls. As expected, defense response was the highest enriched biological process in the DEGs, with six genes related to immune response against pathogens (CHI3L1, SLPI, ACOD1, CCL5, MPO, BPI) included among the ten most expressed genes; and two of the key co-expression modules were associated with regulation of immune response, which also positively correlated with clinical stage and blood monocyte concentration. In particular, sick dogs displayed significant changes in the expression of Th1, Th2, Th17 and Tr1 cytokines (e. g. TNF-α, IFN-γ, IL-21, IL-17, IL-15), markers of T cell and NK cell exhaustion (e. g. LAG3, CD244, Blimp-1, JUN), and B cell, monocyte and macrophage disrupted functionality (e. g. CD40LG, MAPK4, IL-1R, NLRP3, BCMA). In addition, we found an overexpression of XBP1 and some other genes involved in endoplasmic reticulum stress and the IRE1 branch of the unfolded protein response, as well as one co-expression module associated with these processes, which could be induced by L. infantum to prevent host cell apoptosis and modulate inflammation-induced lymphangiogenesis at lymph nodes. Moreover, 21 lncRNAs were differentially expressed in sick dogs, and one key co-expression module was associated with chromatin organization, suggesting that epigenetic mechanisms could also contribute to dampening host immune response during natural L. infantum infection in the lymph nodes of dogs suffering from clinical leishmaniosis.
Collapse
Affiliation(s)
- Carolina R Sanz
- Animal Health Department, Veterinary Faculty, Complutense University of Madrid, Madrid, Spain
| | - Guadalupe Miró
- Animal Health Department, Veterinary Faculty, Complutense University of Madrid, Madrid, Spain
| | - Natalia Sevane
- Department of Animal Production, Veterinary Faculty, Complutense University of Madrid, Madrid, Spain
| | - Armando Reyes-Palomares
- Department of Biochemistry and Molecular Biology, Complutense University of Madrid, Madrid, Spain
| | - Susana Dunner
- Department of Animal Production, Veterinary Faculty, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
12
|
Häfner S. Polymeric Promotion. Microbes Infect 2021; 24:104910. [PMID: 34838737 DOI: 10.1016/j.micinf.2021.104910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 11/24/2022]
Affiliation(s)
- Sophia Häfner
- University of Copenhagen, BRIC Biotech Research & Innovation Centre, Lund Group, 2200 Copenhagen, Denmark.
| |
Collapse
|
13
|
Volpedo G, Pacheco-Fernandez T, Bhattacharya P, Oljuskin T, Dey R, Gannavaram S, Satoskar AR, Nakhasi HL. Determinants of Innate Immunity in Visceral Leishmaniasis and Their Implication in Vaccine Development. Front Immunol 2021; 12:748325. [PMID: 34712235 PMCID: PMC8546207 DOI: 10.3389/fimmu.2021.748325] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/27/2021] [Accepted: 09/24/2021] [Indexed: 12/22/2022] Open
Abstract
Leishmaniasis is endemic to the tropical and subtropical regions of the world and is transmitted by the bite of an infected sand fly. The multifaceted interactions between Leishmania, the host innate immune cells, and the adaptive immunity determine the severity of pathogenesis and disease development. Leishmania parasites establish a chronic infection by subversion and attenuation of the microbicidal functions of phagocytic innate immune cells such as neutrophils, macrophages and dendritic cells (DCs). Other innate cells such as inflammatory monocytes, mast cells and NK cells, also contribute to resistance and/or susceptibility to Leishmania infection. In addition to the cytokine/chemokine signals from the innate immune cells, recent studies identified the subtle shifts in the metabolic pathways of the innate cells that activate distinct immune signal cascades. The nexus between metabolic pathways, epigenetic reprogramming and the immune signaling cascades that drive the divergent innate immune responses, remains to be fully understood in Leishmania pathogenesis. Further, development of safe and efficacious vaccines against Leishmaniasis requires a broader understanding of the early interactions between the parasites and innate immune cells. In this review we focus on the current understanding of the specific role of innate immune cells, the metabolomic and epigenetic reprogramming and immune regulation that occurs during visceral leishmaniasis, and the strategies used by the parasite to evade and modulate host immunity. We highlight how such pathways could be exploited in the development of safe and efficacious Leishmania vaccines.
Collapse
Affiliation(s)
- Greta Volpedo
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Thalia Pacheco-Fernandez
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Parna Bhattacharya
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Timur Oljuskin
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Ranadhir Dey
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Sreenivas Gannavaram
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Abhay R Satoskar
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Hira L Nakhasi
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
14
|
Kar A, Charan Raja MR, Jayaraman A, Srinivasan S, Debnath J, Kar Mahapatra S. Oral combination of eugenol oleate and miltefosine induce immune response during experimental visceral leishmaniasis through nitric oxide generation with advanced cytokine demand. Cytokine 2021; 146:155623. [PMID: 34144446 DOI: 10.1016/j.cyto.2021.155623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/03/2021] [Revised: 06/01/2021] [Accepted: 06/08/2021] [Indexed: 11/24/2022]
Abstract
Conventional therapy of visceral leishmaniasis (VL) remains challenging with the pitfall of toxicity, drug resistance, and expensive. Hence, urgent need for an alternative approach is essential. In this study, we evaluated the potential of combination therapy with eugenol oleate and miltefosine in Leishmania donovani infected macrophages and in the BALB/c mouse model. The interactions between eugenol oleate and miltefosine were found to be additive against promastigotes and amastigotes with xΣFIC 1.13 and 0.68, respectively. Significantly (p < 0.001) decreased arginase activity, increased nitrite generation, improved pro-inflammatory cytokines, and phosphorylated p38MAPK were observed after combination therapy with eugenol oleate and miltefosine. >80% parasite clearance in splenic and hepatic tissue with concomitant nitrite generation, and anti-VL cytokines productions were observed after orally administered miltefosine (5 mg/kg body weight) and eugenol oleate (15 mg/kg body weight) in L. donovani-infected BALB/c mice. Altogether, this study suggested the possibility of an oral combination of miltefosine with eugenol oleate against visceral leishmaniasis.
Collapse
Affiliation(s)
- Amrita Kar
- Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Mamilla R Charan Raja
- Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India; Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai 600077, India
| | - Adithyan Jayaraman
- Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Sujatha Srinivasan
- Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Joy Debnath
- Department of Chemistry, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Santanu Kar Mahapatra
- Department of Biotechnology, School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India; Department of Paramedical and Allied Health Sciences, Midnapore City College, Midnapore 721129, West Bengal, India.
| |
Collapse
|
15
|
Preclinical validation of a live attenuated dermotropic Leishmania vaccine against vector transmitted fatal visceral leishmaniasis. Commun Biol 2021; 4:929. [PMID: 34330999 PMCID: PMC8324786 DOI: 10.1038/s42003-021-02446-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/24/2021] [Accepted: 07/07/2021] [Indexed: 01/06/2023] Open
Abstract
Visceral Leishmaniasis (VL), a potentially fatal disease is caused by Leishmania donovani parasites with no vaccine available. Here we produced a dermotropic live attenuated centrin gene deleted Leishmania major (LmCen−/−) vaccine under Good Laboratory Practices and demonstrated that a single intradermal injection confers robust and durable protection against lethal VL transmitted naturally via bites of L. donovani-infected sand flies and prevents mortality. Surprisingly, immunogenicity characteristics of LmCen−/− parasites revealed activation of common immune pathways like L. major wild type parasites. Spleen cells from LmCen−/− immunized and L. donovani challenged hamsters produced significantly higher Th1-associated cytokines including IFN-γ, TNF-α, and reduced expression of the anti-inflammatory cytokines like IL-10, IL-21, compared to non-immunized challenged animals. PBMCs, isolated from healthy people from non-endemic region, upon LmCen−/− infection also induced more IFN-γ compared to IL-10, consistent with our immunogenicity data in LmCen−/− immunized hamsters. This study demonstrates that the LmCen−/− parasites are safe and efficacious against VL and is a strong candidate vaccine to be tested in a human clinical trial. Karmakar et al produced a dermotropic, live attenuated centrin gene-deleted Leishmania major (LmCen−/−) vaccine against Visceral Leishmaniasis (VL). They demonstrated in hamsters that a single intradermal injection confers robust and durable protection against lethal VL that is transmitted naturally via bites of L. donovani-infected sand flies.
Collapse
|
16
|
Shukla D, Patidar A, Sarma U, Chauhan P, Pandey SP, Chandel HS, Bodhale N, Ghosh SK, Guzman CA, Ebensen T, Silvestre R, Sarkar A, Saha B, Bhattacharjee S. Interdependencies between Toll-like receptors in Leishmania infection. Immunology 2021; 164:173-189. [PMID: 33964011 DOI: 10.1111/imm.13364] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/03/2021] [Accepted: 04/21/2021] [Indexed: 12/22/2022] Open
Abstract
Multiple pathogen-associated molecular patterns (PAMPs) on a pathogen's surface imply their simultaneous recognition by the host cell membrane-located multiple PAMP-specific Toll-like receptors (TLRs). The TLRs on endosomes recognize internalized pathogen-derived nucleic acids and trigger anti-pathogen immune responses aimed at eliminating the intracellular pathogen. Whether the TLRs influence each other's expression and effector responses-termed TLR interdependency-remains unknown. Herein, we first probed the existence of TLR interdependencies and next determined how targeting TLR interdependencies might determine the outcome of Leishmania infection. We observed that TLRs selectively altered expression of their own and of other TLRs revealing novel TLR interdependencies. Leishmania major-an intra-macrophage parasite inflicting the disease cutaneous leishmaniasis in 88 countries-altered this TLR interdependency unfolding a unique immune evasion mechanism. We targeted this TLR interdependency by selective silencing of rationally chosen TLRs and by stimulation with selective TLR ligands working out a novel phase-specific treatment regimen. Targeting the TLR interdependency elicited a host-protective anti-leishmanial immune response and reduced parasite burden. To test whether this observation could be used as a scientific rationale for treating a potentially fatal L. donovani infection, which causes visceral leishmaniasis, we targeted the inter-TLR dependency adopting the same treatment regimen. We observed reduced splenic Leishman-Donovan units accompanied by host-protective immune response in susceptible BALB/c mice. The TLR interdependency optimizes TLR-induced immune response by a novel immunoregulatory framework and scientifically rationalizes targeting TLRs in tandem and in sequence for redirecting immune responses against an intracellular pathogen.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Neelam Bodhale
- Jagadis Bose National Science Talent Search, Kolkata, India
| | | | | | - Thomas Ebensen
- Helmholtz Center for Infectious Diseases, Braunschweig, Germany
| | | | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar, India
| | - Bhaskar Saha
- National Centre for Cell Science, Pune, India.,Trident Academy of Creative Technology, Bhubaneswar, India
| | | |
Collapse
|
17
|
Cortés-Vieyra R, Silva-García O, Gómez-García A, Gutiérrez-Castellanos S, Álvarez-Aguilar C, Baizabal-Aguirre VM. Glycogen Synthase Kinase 3β Modulates the Inflammatory Response Activated by Bacteria, Viruses, and Parasites. Front Immunol 2021; 12:675751. [PMID: 34017345 PMCID: PMC8129516 DOI: 10.3389/fimmu.2021.675751] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/03/2021] [Accepted: 04/12/2021] [Indexed: 01/12/2023] Open
Abstract
Knowledge of glycogen synthase kinase 3β (GSK3β) activity and the molecules identified that regulate its function in infections caused by pathogenic microorganisms is crucial to understanding how the intensity of the inflammatory response can be controlled in the course of infections. In recent years many reports have described small molecular weight synthetic and natural compounds, proteins, and interference RNA with the potential to regulate the GSK3β activity and reduce the deleterious effects of the inflammatory response. Our goal in this review is to summarize the most recent advances on the role of GSK3β in the inflammatory response caused by bacteria, bacterial virulence factors (i.e. LPS and others), viruses, and parasites and how the regulation of its activity, mainly its inhibition by different type of molecules, modulates the inflammation.
Collapse
Affiliation(s)
- Ricarda Cortés-Vieyra
- División de Investigación Clínica, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social (IMSS), Morelia, Mexico
| | - Octavio Silva-García
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Anel Gómez-García
- División de Investigación Clínica, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social (IMSS), Morelia, Mexico
| | - Sergio Gutiérrez-Castellanos
- División de Investigación Clínica, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social (IMSS), Morelia, Mexico
| | - Cleto Álvarez-Aguilar
- Coordinación Auxiliar Médica de Investigación en Salud, IMSS Michoacán, Morelia, Mexico
| | - Víctor M Baizabal-Aguirre
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| |
Collapse
|
18
|
Bamra T, Shafi T, Das S, Kumar M, Dikhit MR, Kumar A, Kumar A, Abhishek K, Pandey K, Sen A, Das P. Leishmania donovani Secretory Mevalonate Kinase Regulates Host Immune Response and Facilitates Phagocytosis. Front Cell Infect Microbiol 2021; 11:641985. [PMID: 33981628 PMCID: PMC8110032 DOI: 10.3389/fcimb.2021.641985] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/15/2020] [Accepted: 03/31/2021] [Indexed: 11/13/2022] Open
Abstract
Leishmania secretes over 151 proteins during in vitro cultivation. Cellular functions of one such novel protein: mevalonate kinase is discussed here; signifying its importance in Leishmania infection. Visceral Leishmaniasis is a persistent infection, caused by Leishmania donovani in Indian subcontinent. This persistence is partly due to phagocytosis and evasion of host immune response. The underlying mechanism involves secretory proteins of Leishmania parasite; however, related studies are meagre. We have identified a novel secretory Leishmania donovani glycoprotein, Mevalonate kinase (MVK), and shown its importance in parasite internalization and immuno-modulation. In our studies, MVK was found to be secreted maximum after 1 h temperature stress at 37°C. Its secretion was increased by 6.5-fold in phagolysosome-like condition (pH ~5.5, 37°C) than at pH ~7.4 and 25°C. Treatment with MVK modulated host immune system by inducing interleukin-10 and interleukin-4 secretion, suppressing host’s ability to kill the parasite. Peripheral blood mononuclear cell (PBMC)-derived macrophages infected with mevalonate kinase-overexpressing parasites showed an increase in intracellular parasite burden in comparison to infection with vector control parasites. Mechanism behind the increase in phagocytosis and immunosuppression was found to be phosphorylation of mitogen-activated protein (MAP) kinase pathway protein, Extracellular signal-regulated kinases-1/2, and actin scaffold protein, cortactin. Thus, we conclude that Leishmania donovani Mevalonate kinase aids in parasite engulfment and subvert the immune system by interfering with signal transduction pathways in host cells, which causes suppression of the protective response and facilitates their persistence in the host. Our work elucidates the involvement of Leishmania in the process of phagocytosis which is thought to be dependent largely on macrophages and contributes towards better understanding of host pathogen interactions.
Collapse
Affiliation(s)
- Tanvir Bamra
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Taj Shafi
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | | | - Manjay Kumar
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Manas Ranjan Dikhit
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Ajay Kumar
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Ashish Kumar
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Kumar Abhishek
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Krishna Pandey
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Abhik Sen
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | - Pradeep Das
- Department of Molecular Biology, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| |
Collapse
|
19
|
Ulusan Ö, Mert U, Sadıqova A, Öztürk S, Caner A. Identification of gene expression profiles in Leishmania major infection by integrated bioinformatics analyses. Acta Trop 2020; 208:105517. [PMID: 32360239 DOI: 10.1016/j.actatropica.2020.105517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/27/2020] [Revised: 04/22/2020] [Accepted: 04/22/2020] [Indexed: 10/24/2022]
Abstract
Gene expression profiling in mouse models of leishmaniasis has given useful information to understand the molecular pathways active in lesions and to discover new diagnostic/therapeutic targets. Although the host response plays a critical role in protection from leishmaniasis and promoting disease severity, there are still unexplained aspects in the mechanism of non-healing cutaneous lesions, which need biomarkers for both targeted- therapy and diagnosis. To address this, transcriptional profiling of the skin lesions obtained from BALB/c mice infected with Leishmania major and healthy skin from naïve mice were evaluated by bioinformatics analysis, and then the results were validated by Revers Transcriptase-PCR. Five genes among the up-regulated differentially expressed genes named FCGR4, CCL4, CXCL9, Arg1 and IL-1β were found to have relatively high diagnostic value for CL due to L. major. Pathway analysis revealed that Triggering Receptor Expressed On Myeloid Cells 1 (TREM1) signaling pathways are active in cutaneous lesions, providing new insights for the understanding and treatment of leishmaniasis.
Collapse
Affiliation(s)
- Özlem Ulusan
- Department of Parasitology, Ege University Medical School, Izmir, Turkey
| | - Ufuk Mert
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Aygül Sadıqova
- Infectious Disease Division, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Sercan Öztürk
- Departments of Computer Science, Dokuz Eylül University, Izmir, Turkey
| | - Ayse Caner
- Department of Parasitology, Ege University Medical School, Izmir, Turkey; Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey; Department of Bioinformatics, Ege University, Institute of Health Sciences, Izmir, Turkey; Departments of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
20
|
Jha MK, Sarode AY, Bodhale N, Mukherjee D, Pandey SP, Srivastava N, Rub A, Silvestre R, Sarkar A, Saha B. Development and Characterization of an Avirulent Leishmania major Strain. THE JOURNAL OF IMMUNOLOGY 2020; 204:2734-2753. [PMID: 32245818 DOI: 10.4049/jimmunol.1901362] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 11/14/2019] [Accepted: 03/05/2020] [Indexed: 01/12/2023]
Abstract
Leishmania major causes cutaneous leishmaniasis. An antileishmanial vaccine for humans is unavailable. In this study, we report development of two attenuated L. major strains-5ASKH-HP and LV39-HP-by continuous culture (high passage) of the corresponding virulent strains (low passage). Both avirulent strains showed similar changes in proteome profiles when analyzed by surface-enhanced laser desorption ionization mass spectrometry. Liquid chromatography-mass spectrometry and microarray characterization of 5ASKH strains revealed substantially altered gene and protein expression profiles, respectively. Both virulent and avirulent L. major strains grew comparably in culture, but the avirulent strain survived significantly less in BALB/c-derived peritoneal macrophages. Both attenuated strains failed to infect BALB/c mice and elicited IFN-γ, but not IL-4 and IL-10, responses. 5ASKH-HP parasites failed to induce significant infection even in severely immunocompromised- SCID or inducible NO synthase-, CD40-, or IL-12-deficient mice, indicating attenuation. The avirulent strain induced less IL-10, but higher IL-12, in macrophages. The avirulent strain failed to reduce CD40 relocation to the detergent-resistant membrane domain and to inhibit CD40-induced phosphorylation of the kinases Lyn and protein kinase C-β and MAPKs MKK-3/6 and p38MAPK or to upregulate MEK-1/2 and ERK-1/2 in BALB/c-derived peritoneal macrophages. The virulent and the avirulent strains reciprocally modulated CD40-induced Ras-mediated signaling through PI-3K and Raf-1. Avirulent 5ASKH-primed BALB/c mice were protected against virulent L. major challenge infection. The loss of virulence accompanied by substantially altered proteome profiles and the elicitation of host-protective immune responses indicate plausibly irreversible attenuation of the L. major strain and its potential use as a vaccine strain.
Collapse
Affiliation(s)
- Mukesh Kumar Jha
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, Maharashtra 411007, India
| | - Aditya Y Sarode
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, Maharashtra 411007, India
| | - Neelam Bodhale
- Jagadis Bose National Science Talent Search, Kolkata, West Bengal 700107, India
| | - Debasri Mukherjee
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, Maharashtra 411007, India
| | - Surya Prakash Pandey
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, Maharashtra 411007, India
| | - Neetu Srivastava
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, Maharashtra 411007, India
| | - Abdur Rub
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, Maharashtra 411007, India
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, 4710-057 Braga, Portugal; and
| | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar, Odisha 751024, India
| | - Bhaskar Saha
- National Centre for Cell Science, Savitribai Phule Pune University, Pune, Maharashtra 411007, India; .,Trident Academy of Creative Technology, Bhubaneswar, Odisha 751024, India
| |
Collapse
|
21
|
Pradhan G, Raj Abraham P, Shrivastava R, Mukhopadhyay S. Calcium Signaling Commands Phagosome Maturation Process. Int Rev Immunol 2020; 38:57-69. [PMID: 31117900 DOI: 10.1080/08830185.2019.1592169] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/17/2022]
Abstract
Phagosome-lysosome (P-L) fusion is one of the central immune-effector responses of host. It is known that phagosome maturation process is associated with numerous signaling cascades and among these, important role of calcium (Ca2+) signaling has been realized recently. Ca2+ plays key roles in actin rearrangement, activation of NADPH oxidase and protein kinase C (PKC). Involvement of Ca2+ in these cellular processes directs phagosomal maturation process. Some of the intracellular pathogens have acquired the strategies to modulate Ca2+ associated pathways to block P-L fusion process. In this review we have described the mechanism of Ca2+ signals that influence P-L fusion by controlling ROS, actin and PKC signaling cascades. We have also discussed the strategies implemented by the intracellular pathogens to manipulate Ca2+ signaling to consequently subvert P-L fusion. A detail study of factors associated in manipulating Ca2+ signaling may provide new insights for the development of therapeutic tools for more effective treatment options against infectious diseases.
Collapse
Affiliation(s)
- Gourango Pradhan
- a Laboratory of Molecular Cell Biology , Centre for DNA Fingerprinting and Diagnostics (CDFD) , Hyderabad , India.,b Graduate Studies , Manipal Academy of Higher Education , Manipal , Karnataka , India
| | - Philip Raj Abraham
- a Laboratory of Molecular Cell Biology , Centre for DNA Fingerprinting and Diagnostics (CDFD) , Hyderabad , India
| | - Rohini Shrivastava
- a Laboratory of Molecular Cell Biology , Centre for DNA Fingerprinting and Diagnostics (CDFD) , Hyderabad , India.,b Graduate Studies , Manipal Academy of Higher Education , Manipal , Karnataka , India
| | - Sangita Mukhopadhyay
- a Laboratory of Molecular Cell Biology , Centre for DNA Fingerprinting and Diagnostics (CDFD) , Hyderabad , India
| |
Collapse
|
22
|
Banerjee S, Mukherjee N, Gajbhiye RL, Mishra S, Jaisankar P, Datta S, Das Saha K. Intracellular anti-leishmanial effect of Spergulin-A, a triterpenoid saponin of Glinus oppositifolius. Infect Drug Resist 2019; 12:2933-2942. [PMID: 31571946 PMCID: PMC6756365 DOI: 10.2147/idr.s211721] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/09/2019] [Accepted: 08/06/2019] [Indexed: 12/19/2022] Open
Abstract
Background Many of present chemotherapeutics are inadequate and also resistant against visceral leishmaniasis (VL), an immunosuppressive ailment caused by Leishmania donovani. Despite the interest in plant-based drug development, no antileishmanial drugs from plant source are currently available. Glinus oppositifolius had been reported in favor of being immune modulators along with other traditional uses. Novel anti-VL therapies can rely on host immune-modulation with associated leishmanicidal action. Objective Discovery of novel plant-based antileishmanial compound from G. oppositifolius having permissible side effects. Methods With this rationale, an n-BuOH fraction of the methanolic extract of the plant and obtained triterpenoid saponin Spergulin-A were evaluated against acellular and intracellular L. donovani. Immunostimulatory activity of them was confirmed by elevated TNF-α and extracellular NO production from treated MФs and was found nontoxic to the host cells. Identification and structure confirmation for isolated Spergulin-A was performed by ESI-MS,13C, and 1H NMR. Results Spergulin-A was found ineffective against the acellular forms while, against the intracellular parasites at 30 μg/mL, the reduction was 92.6% after 72 hrs. Spergulin-A enhanced ROS and nitric oxide (NO) release and changes in Gp91-phox, i-NOS, and pro and anti-inflammatory cytokines elaborated its intracellular anti-leishmanial activity. Conclusion The results supported that G. oppositifolius and Spergulin-A can potentiate new lead molecules for the development of alternative drugs against VL.
Collapse
Affiliation(s)
- Saswati Banerjee
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Niladri Mukherjee
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Rahul L Gajbhiye
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Snehasis Mishra
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Parasuraman Jaisankar
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Sriparna Datta
- Department of Chemical Technology, University of Calcutta, Kolkata 700009, India
| | - Krishna Das Saha
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| |
Collapse
|
23
|
Systematic Review of Host-Mediated Activity of Miltefosine in Leishmaniasis through Immunomodulation. Antimicrob Agents Chemother 2019; 63:AAC.02507-18. [PMID: 31036692 PMCID: PMC6591591 DOI: 10.1128/aac.02507-18] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/03/2018] [Accepted: 04/22/2019] [Indexed: 12/12/2022] Open
Abstract
Host immune responses are pivotal for the successful treatment of the leishmaniases, a spectrum of infections caused by Leishmania parasites. Previous studies speculated that augmenting cytokines associated with a type 1 T-helper cell (Th1) response is necessary to combat severe forms of leishmaniasis, and it has been hypothesized that the antileishmanial drug miltefosine is capable of immunomodulation and induction of Th1 cytokines. Host immune responses are pivotal for the successful treatment of the leishmaniases, a spectrum of infections caused by Leishmania parasites. Previous studies speculated that augmenting cytokines associated with a type 1 T-helper cell (Th1) response is necessary to combat severe forms of leishmaniasis, and it has been hypothesized that the antileishmanial drug miltefosine is capable of immunomodulation and induction of Th1 cytokines. A better understanding of the immunomodulatory effects of miltefosine is central to providing a rationale regarding synergistic mechanisms of activity to combine miltefosine optimally with other conventional and future antileishmanials that are currently under development. Therefore, a systematic literature search was performed to evaluate to what extent and how miltefosine influences the host Th1 response. Miltefosine’s effects observed in both a preclinical and a clinical context associated with immunomodulation in the treatment of leishmaniasis are evaluated in this review. A total of 27 studies were included in the analysis. Based on the current evidence, miltefosine is not only capable of inducing direct parasite killing but also of modulating the host immunity. Our findings suggest that miltefosine-induced activation of Th1 cytokines, particularly represented by increased gamma interferon (IFN-γ) and interleukin 12 (IL-12), is essential to prevail over the Leishmania-driven Th2 response. Differences in miltefosine-induced host-mediated effects between in vitro, ex vivo, animal model, and human studies are further discussed. All things considered, an effective treatment with miltefosine is acquired by enhanced functional Th1 cytokine responses and may further be enhanced in combination with immunostimulatory agents.
Collapse
|
24
|
Saha S, Basu M, Guin S, Gupta P, Mitterstiller AM, Weiss G, Jana K, Ukil A. Leishmania donovani Exploits Macrophage Heme Oxygenase-1 To Neutralize Oxidative Burst and TLR Signaling-Dependent Host Defense. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 202:827-840. [PMID: 30593539 DOI: 10.4049/jimmunol.1800958] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 07/09/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
Suppression of host oxidative burst is essential for survival of the intracellular parasite Leishmania donovani Screening of macrophage antioxidant enzymes during infection revealed marked upregulation of the heme-degrading enzyme, heme oxygenase-1 (HO-1). Moreover, HO-1-silenced RAW macrophages depicted increased superoxide production and decreased parasite survival. HO-1 induction decreased cellular heme content, thereby inhibiting the heme-dependent maturation of gp91phox, a catalytic component of major reactive oxygen species-producing enzyme NAD(P)H oxidase. Decreased gp91phox expression resulted in reduced stability of p22phox, another component of the catalytic center of NAD(P)H oxidase. Replenishing infected cells with exogenous heme reversed these effects and restored NAD(P)H oxidase activity. Persistent HO-1 expression at late hour of infection prompted us to investigate its effect on other host defense parameters, and inhibition study revealed a reciprocal relationship of HO-1 with host proinflammatory responses. Among all the HO-1-mediated heme degradation products (CO, Fe, and biliverdin), only CO documented potent anti-inflammatory effects. Quenching of CO during infection increased the production of disease-resolving cytokines IL-12 and TNF-α. Coimmunoprecipitation experiments revealed that CO inhibited the interaction of TLR4 with MyD88 and TIR domain-containing adapter-inducing IFN-β, thereby dampening the activation of NF-κB and IFN regulatory factor 3-mediated production of proinflammatory cytokines. Administration of HO-1 inhibitor tin protoporphyrin IX dichloride in infected BALB/c mice led to a decrease in liver and spleen parasite burden along with increased production of IL-12 and TNF-α. These results suggest that HO-1 on one hand inhibits reactive oxygen species generation and on the other hand downregulates host favorable cytokine responses, thereby facilitating intramacrophage parasite survival.
Collapse
Affiliation(s)
- Shriya Saha
- Department of Biochemistry, University of Calcutta, Kolkata 700019, India
| | - Moumita Basu
- Department of Biochemistry, University of Calcutta, Kolkata 700019, India
| | - Subham Guin
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata 700032, India
| | - Purnima Gupta
- Infections and Cancer Biology Group, International Agency for Research on Cancer, 69372 Lyon Cedex 08, France
| | - Anna-Maria Mitterstiller
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, 69008 Innsbruck, Austria; and
| | - Guenter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Pneumology, Medical University of Innsbruck, 69008 Innsbruck, Austria; and
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, Kolkata 700054, India
| | - Anindita Ukil
- Department of Biochemistry, University of Calcutta, Kolkata 700019, India;
| |
Collapse
|
25
|
HIF-1α-regulated MIF activation and Nox2-dependent ROS generation promote Leishmania amazonensis killing by macrophages under hypoxia. Cell Immunol 2019; 335:15-21. [DOI: 10.1016/j.cellimm.2018.10.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/12/2018] [Revised: 10/08/2018] [Accepted: 10/21/2018] [Indexed: 12/12/2022]
|
26
|
Edwardsiella piscicida Type III Secretion System Effector EseK Inhibits Mitogen-Activated Protein Kinase Phosphorylation and Promotes Bacterial Colonization in Zebrafish Larvae. Infect Immun 2018; 86:IAI.00233-18. [PMID: 29986890 DOI: 10.1128/iai.00233-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/03/2018] [Accepted: 07/01/2018] [Indexed: 12/21/2022] Open
Abstract
Bacteria utilize type III secretion systems (T3SS) to deliver effectors directly into host cells. Hence, it is very important to identify the functions of bacterial (T3SS) effectors to understand host-pathogen interactions. Edwardsiella piscicida encodes a functional T3SS effector, EseK, which can be translocated into host cells and affect bacterial loads. Here, it was demonstrated that an eseK mutant (the ΔeseK mutant) significantly increased the phosphorylation levels of p38α, c-Jun NH2-terminal kinases (JNK), and extracellular signal-regulated protein kinases 1/2 (ERK1/2) in HeLa cells. Overexpression of EseK directly inhibited mitogen-activated protein kinase (MAPK) signaling pathways in HEK293T cells. The ΔeseK mutant consistently promoted the phosphorylation of MAPKs in zebrafish larva infection models. Further, it was shown that the ΔeseK mutant increased the expression of tumor necrosis factor alpha (TNF-α) in an MAPK-dependent manner. Importantly, the EseK-mediated inhibition of MAPKs in vivo attenuated bacterial clearance in larvae. Taken together, this work reveals that the E. piscicida T3SS effector EseK promotes bacterial infection by inhibiting MAPK activation, which provides insights into the molecular pathogenesis of E. piscicida in fish.
Collapse
|
27
|
Husein A, Jamal A, Ahmed MZ, Arish M, Ali R, Tabrez S, Rasool F, Rub A. Leishmania donovani infection differentially regulates small G-proteins. J Cell Biochem 2018; 119:7844-7854. [PMID: 29943842 DOI: 10.1002/jcb.27186] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/11/2018] [Accepted: 05/24/2018] [Indexed: 01/28/2023]
Abstract
Leishmania is a protozoan parasite that resides and replicates in macrophages and causes leishmaniasis. The parasite alters the signaling cascade in host macrophages and evades the host machinery. Small G-proteins are GTPases, grouped in 5 different families that play a crucial role in the regulation of cell proliferation, cell survival, apoptosis, intracellular trafficking, and transport. In particular, the Ras family of small G-proteins has been identified to play a significant role in the cellular functions mentioned before. Here, we studied the differential expression of the most important small G-proteins during Leishmania infection. We found major changes in the expression of different isoforms of Ras, mainly in N-Ras. We observed that Leishmania donovani infection led to enhanced N-Ras expression, whereas it inhibited K-Ras and H-Ras expression. Furthermore, an active N-Ras pull-down assay showed enhanced N-Ras activity. L donovani infection also increased extracellular signal-regulated kinase 1/2 phosphorylation and simultaneously decreased p38 phosphorylation. In contrast, pharmacological inhibition of Ras led to reduction in the phosphorylation of extracellular signal-regulated kinase 1/2 and enhanced the phosphorylation of p38 in Leishmania-infected cells, which could lead to increased interleukin-12 expression and decreased interleukin-10 expression. Indeed, farnesylthiosalicyclic acid (a Ras inhibitor), when used at the effective level in L donovani-infected macrophages, reduced amastigotes in the host macrophages. Thus, upregulated N-Ras expression during L donovani infection could be a novel immune evasion strategy of Leishmania and would be a potential target for antileishmanial immunotherapy.
Collapse
Affiliation(s)
- Atahar Husein
- Infection and Immunity Lab (Lab No. 414), Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Azfar Jamal
- Virology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Mohammad Zulfazal Ahmed
- Infection and Immunity Lab (Lab No. 414), Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Mohammad Arish
- Infection and Immunity Lab (Lab No. 414), Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Rahat Ali
- Infection and Immunity Lab (Lab No. 414), Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Shams Tabrez
- Infection and Immunity Lab (Lab No. 414), Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Fayyaz Rasool
- Infection and Immunity Lab (Lab No. 414), Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Abdur Rub
- Infection and Immunity Lab (Lab No. 414), Department of Biotechnology, Jamia Millia Islamia, New Delhi, India.,Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
| |
Collapse
|
28
|
Queval CJ, Song OR, Carralot JP, Saliou JM, Bongiovanni A, Deloison G, Deboosère N, Jouny S, Iantomasi R, Delorme V, Debrie AS, Park SJ, Gouveia JC, Tomavo S, Brosch R, Yoshimura A, Yeramian E, Brodin P. Mycobacterium tuberculosis Controls Phagosomal Acidification by Targeting CISH-Mediated Signaling. Cell Rep 2018; 20:3188-3198. [PMID: 28954234 PMCID: PMC5637157 DOI: 10.1016/j.celrep.2017.08.101] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/28/2017] [Revised: 07/29/2017] [Accepted: 08/30/2017] [Indexed: 11/29/2022] Open
Abstract
Pathogens have evolved a range of mechanisms to counteract host defenses, notably to survive harsh acidic conditions in phagosomes. In the case of Mycobacterium tuberculosis, it has been shown that regulation of phagosome acidification could be achieved by interfering with the retention of the V-ATPase complexes at the vacuole. Here, we present evidence that M. tuberculosis resorts to yet another strategy to control phagosomal acidification, interfering with host suppressor of cytokine signaling (SOCS) protein functions. More precisely, we show that infection of macrophages with M. tuberculosis leads to granulocyte-macrophage colony-stimulating factor (GM-CSF) secretion, inducing STAT5-mediated expression of cytokine-inducible SH2-containing protein (CISH), which selectively targets the V-ATPase catalytic subunit A for ubiquitination and degradation by the proteasome. Consistently, we show that inhibition of CISH expression leads to reduced replication of M. tuberculosis in macrophages. Our findings further broaden the molecular understanding of mechanisms deployed by bacteria to survive. M. tuberculosis interferes with host pathways to control vacuolar acidification Infection induces the expression of host CISH and recruitment to the phagosome CISH triggers the degradation of H+-V-ATPase via SOCS box-mediated ubiquitination This defense mechanism complements previous schemes relying on virulence factors
Collapse
Affiliation(s)
- Christophe J Queval
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204, CIIL-Center for Infection and Immunity of Lille, 59000 Lille, France; Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics, 75015 Paris, France
| | - Ok-Ryul Song
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204, CIIL-Center for Infection and Immunity of Lille, 59000 Lille, France; Institut Pasteur Korea, 16 Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, South Korea
| | - Jean-Philippe Carralot
- Institut Pasteur Korea, 16 Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, South Korea
| | - Jean-Michel Saliou
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204, CIIL-Center for Infection and Immunity of Lille, 59000 Lille, France; Plateforme de Protéomique et Peptides Modifiés (P3M), CNRS, Institut Pasteur de Lille, University Lille, 59000 Lille, France
| | - Antonino Bongiovanni
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204, CIIL-Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Gaspard Deloison
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204, CIIL-Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Nathalie Deboosère
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204, CIIL-Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Samuel Jouny
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204, CIIL-Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Raffaella Iantomasi
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204, CIIL-Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Vincent Delorme
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204, CIIL-Center for Infection and Immunity of Lille, 59000 Lille, France; Institut Pasteur Korea, 16 Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, South Korea
| | - Anne-Sophie Debrie
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204, CIIL-Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Sei-Jin Park
- Institut Pasteur Korea, 16 Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, South Korea
| | - Joana Costa Gouveia
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204, CIIL-Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Stanislas Tomavo
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204, CIIL-Center for Infection and Immunity of Lille, 59000 Lille, France; Plateforme de Protéomique et Peptides Modifiés (P3M), CNRS, Institut Pasteur de Lille, University Lille, 59000 Lille, France
| | - Roland Brosch
- Institut Pasteur, Unit for Integrated Mycobacterial Pathogenomics, 75015 Paris, France
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo 160-8582, Japan
| | - Edouard Yeramian
- Unité de Microbiologie Structurale, CNRS UMR3528 Institut Pasteur, 75015 Paris, France.
| | - Priscille Brodin
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204, CIIL-Center for Infection and Immunity of Lille, 59000 Lille, France; Institut Pasteur Korea, 16 Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, South Korea.
| |
Collapse
|
29
|
Azami M, Ranjkesh Adermanabadi V, Khanahmad H, Mohaghegh MA, Zaherinejad E, Aghaei M, Jalali A, Hejazi SH. Immunology and Genetic of Leishmania infantum: The Role of Endonuclease G in the Apoptosis. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2018; 23:36. [PMID: 29887904 PMCID: PMC5961285 DOI: 10.4103/jrms.jrms_705_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Academic Contribution Register] [Received: 09/04/2017] [Revised: 01/13/2018] [Accepted: 01/31/2018] [Indexed: 12/13/2022]
Abstract
Leishmania infantum is the causative agent of infantile visceral leishmaniasis (VL) in the Mediterranean region. Despite developing protective responses, the disease progresses due to many of factors. These include the action of suppressive cytokines, exhaustion of specific T cells, loss of lymphoid tissue, and defective humoral response. Genetic changes that occur inside the genome of alienated or parasite cells, along with immune responses, play an important role in controlling or progressing the disease. Proapoptotic proteins such as Smac/DIABLO, EndoG, AIF (apoptosis-inducing factor), and cytochrome C are effective in apoptosis. EndoG is a mitochondrion-specific nuclease that translocates to the nucleus during apoptosis. Once released from mitochondria, endoG cleaves chromatin DNA into nucleosomal fragments independently of caspases. Therefore, endoG represents a caspase-independent apoptotic pathway initiated from the mitochondria. A comprehensive understanding of the immune and genetic events that occur during VL is very important for designing immunotherapy strategies and developing effective vaccines for disease prevention. In this review which explained the immunological responses and also the important factors that can contribute to parasite apoptosis and are used in subsequent studies as a target for the preparation of drugs or recombinant vaccines against parasites are briefly reviewed.
Collapse
Affiliation(s)
- Mehdi Azami
- Skin Diseases and Leishmaniasis Research Center, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Hossein Khanahmad
- Department of Molecular Biology and Genetics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Ali Mohaghegh
- Department of Laboratory Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.,Health Sciences Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Ebtesam Zaherinejad
- School of Pharmacy, Department of Health Sciences, Curtin University of Technology, Bentley Campus, Australia
| | - Maryam Aghaei
- Skin Diseases and Leishmaniasis Research Center, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Akram Jalali
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Hossein Hejazi
- Skin Diseases and Leishmaniasis Research Center, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Department of Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
30
|
Mahanta A, Ganguli P, Barah P, Sarkar RR, Sarmah N, Phukan S, Bora M, Baruah S. Integrative Approaches to Understand the Mastery in Manipulation of Host Cytokine Networks by Protozoan Parasites with Emphasis on Plasmodium and Leishmania Species. Front Immunol 2018. [PMID: 29527208 PMCID: PMC5829655 DOI: 10.3389/fimmu.2018.00296] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/04/2023] Open
Abstract
Diseases by protozoan pathogens pose a significant public health concern, particularly in tropical and subtropical countries, where these are responsible for significant morbidity and mortality. Protozoan pathogens tend to establish chronic infections underscoring their competence at subversion of host immune processes, an important component of disease pathogenesis and of their virulence. Modulation of cytokine and chemokine levels, their crosstalks and downstream signaling pathways, and thereby influencing recruitment and activation of immune cells is crucial to immune evasion and subversion. Many protozoans are now known to secrete effector molecules that actively modulate host immune transcriptome and bring about alterations in host epigenome to alter cytokine levels and signaling. The complexity of multi-dimensional events during interaction of hosts and protozoan parasites ranges from microscopic molecular levels to macroscopic ecological and epidemiological levels that includes disrupting metabolic pathways, cell cycle (Toxoplasma and Theileria sp.), respiratory burst, and antigen presentation (Leishmania spp.) to manipulation of signaling hubs. This requires an integrative systems biology approach to combine the knowledge from all these levels to identify the complex mechanisms of protozoan evolution via immune escape during host-parasite coevolution. Considering the diversity of protozoan parasites, in this review, we have focused on Leishmania and Plasmodium infections. Along with the biological understanding, we further elucidate the current efforts in generating, integrating, and modeling of multi-dimensional data to explain the modulation of cytokine networks by these two protozoan parasites to achieve their persistence in host via immune escape during host-parasite coevolution.
Collapse
Affiliation(s)
- Anusree Mahanta
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, India.,Institute of Stem Cell Biology and Regenerative Medicine, Bengaluru, India
| | - Piyali Ganguli
- Chemical Engineering and Process Development, CSIR- National Chemical Laboratory, Pune, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-NCL Campus, Pune, India
| | - Pankaj Barah
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, India
| | - Ram Rup Sarkar
- Chemical Engineering and Process Development, CSIR- National Chemical Laboratory, Pune, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-NCL Campus, Pune, India
| | - Neelanjana Sarmah
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, India
| | - Saurav Phukan
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, India
| | - Mayuri Bora
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, India
| | - Shashi Baruah
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, India
| |
Collapse
|
31
|
Knockdown of Host Antioxidant Defense Genes Enhances the Effect of Glucantime on Intracellular Leishmania braziliensis in Human Macrophages. Antimicrob Agents Chemother 2017; 61:AAC.02099-16. [PMID: 28461312 DOI: 10.1128/aac.02099-16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/29/2016] [Accepted: 04/23/2017] [Indexed: 01/01/2023] Open
Abstract
Leishmaniasis is a neglected tropical disease that affects millions of people worldwide and represents a major public health problem. Information on protein expression patterns and functional roles within the context of Leishmania-infected human monocyte-derived macrophages (MDMs) under drug treatment conditions is essential for understanding the role of these cells in leishmaniasis treatment. We analyzed functional changes in the expression of human MDM genes and proteins during in vitro infection by Leishmania braziliensis and treatment with Glucantime (SbV), using quantitative PCR (qPCR) arrays, Western blotting, confocal microscopy, and small interfering RNA (siRNA) human gene inhibition assays. Comparison of the results from gene transcription and protein expression analyses revealed that glutathione S-transferase π1 (GSTP1), glutamate-cysteine ligase modifier subunit (GCLM), glutathione reductase (GSR), glutathione synthetase (GSS), thioredoxin (TRX), and ATP-binding cassette, subfamily B, member 5 (ABCB5), were strongly upregulated at both the mRNA and protein levels in human MDMs that were infected and treated, compared to the control group. Subcellular localization studies showed a primarily phagolysosomal location for the ABCB5 transporter, indicating that this protein may be involved in the transport of SbV By inducing a decrease in L. braziliensis intracellular survival in THP-1 macrophages, siRNA silencing of GSTP1, GSS, and ABCB5 resulted in an increased leishmanicidal effect of SbV exposure in vitro Our results suggest that human MDMs infected with L. braziliensis and treated with SbV express increased levels of genes participating in antioxidant defense, whereas our functional analyses provide evidence for the involvement of human MDMs in drug detoxification. Therefore, we conclude that GSS, GSTP1, and ABCB5 proteins represent potential targets for enhancing the leishmanicidal activity of Glucantime.
Collapse
|
32
|
Zamora-Chimal J, Hernández-Ruiz J, Becker I. NKT cells in leishmaniasis. Immunobiology 2017; 222:641-646. [DOI: 10.1016/j.imbio.2016.11.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/15/2016] [Accepted: 11/30/2016] [Indexed: 10/20/2022]
|
33
|
Geraldo MM, Costa CR, Barbosa FMC, Vivanco BC, Gonzaga WFKM, Novaes E Brito RR, Popi AF, Lopes JD, Xander P. In vivo and in vitro phagocytosis of Leishmania (Leishmania) amazonensis promastigotes by B-1 cells. Parasite Immunol 2017; 38:365-76. [PMID: 27084328 DOI: 10.1111/pim.12324] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/26/2015] [Accepted: 04/06/2016] [Indexed: 12/12/2022]
Abstract
Leishmaniasis is caused by Leishmania parasites that infect several cell types. The promastigote stage of Leishmania is internalized by phagocytic cells and transformed into the obligate intracellular amastigote form. B-1 cells are a subpopulation of B cells that are able to differentiate in vitro and in vivo into mononuclear phagocyte-like cells with phagocytic properties. B-1 cells use several receptors for phagocytosis, such as the mannose receptor and third complement receptor. Leishmania binds to the same receptors on macrophages. In this study, we demonstrated that phagocytes derived from B-1 cells (B-1 CDP) were able to internalize promastigotes of L. (L.) amazonensis in vitro. The internalized promastigotes differentiated into amastigotes. Our results showed that the phagocytic index was higher in B-1 CDP compared to peritoneal macrophages and bone marrow-derived macrophages. The in vivo phagocytic ability of B-1 cells was also demonstrated. Parasites were detected inside purified B-1 cells after intraperitoneal infection with L. (L.) amazonensis promastigotes. Intraperitoneal stimulation with the parasites led to an increase in both IL-10 and TNF-α. These results highlight the importance of studying B-1 CDP cells as phagocytic cells that can participate and contribute to immunity to parasites.
Collapse
Affiliation(s)
- M M Geraldo
- Departamento de Ciências Biológicas, Universidade Federal de São Paulo - Campus Diadema, Diadema, São Paulo, Brazil
| | - C R Costa
- Departamento de Ciências Biológicas, Universidade Federal de São Paulo - Campus Diadema, Diadema, São Paulo, Brazil
| | - F M C Barbosa
- Departamento de Ciências Biológicas, Universidade Federal de São Paulo - Campus Diadema, Diadema, São Paulo, Brazil
| | - B C Vivanco
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - W F K M Gonzaga
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | | | - A F Popi
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - J D Lopes
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil.,Universidade Federal de São Paulo - Campus Diadema, Diadema, São Paulo, Brazil
| | - P Xander
- Departamento de Ciências Biológicas, Universidade Federal de São Paulo - Campus Diadema, Diadema, São Paulo, Brazil
| |
Collapse
|
34
|
Hamidzadeh K, Christensen SM, Dalby E, Chandrasekaran P, Mosser DM. Macrophages and the Recovery from Acute and Chronic Inflammation. Annu Rev Physiol 2017; 79:567-592. [PMID: 27959619 PMCID: PMC5912892 DOI: 10.1146/annurev-physiol-022516-034348] [Citation(s) in RCA: 275] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/11/2022]
Abstract
In recent years, researchers have devoted much attention to the diverse roles of macrophages and their contributions to tissue development, wound healing, and angiogenesis. What should not be lost in the discussions regarding the diverse biology of these cells is that when perturbed, macrophages are the primary contributors to potentially pathological inflammatory processes. Macrophages stand poised to rapidly produce large amounts of inflammatory cytokines in response to danger signals. The production of these cytokines can initiate a cascade of inflammatory mediator release that can lead to wholesale tissue destruction. The destructive inflammatory capability of macrophages is amplified by exposure to exogenous interferon-γ, which prolongs and heightens inflammatory responses. In simple terms, macrophages can thus be viewed as incendiary devices with hair triggers waiting to detonate. We have begun to ask questions about how these cells can be regulated to mitigate the collateral destruction associated with macrophage activation.
Collapse
Affiliation(s)
- Kajal Hamidzadeh
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland 20742;
| | - Stephen M Christensen
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland 20742;
| | - Elizabeth Dalby
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland 20742;
| | - Prabha Chandrasekaran
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland 20742;
| | - David M Mosser
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute, University of Maryland, College Park, Maryland 20742;
| |
Collapse
|
35
|
Gupta P, Srivastav S, Saha S, Das PK, Ukil A. Leishmania donovani inhibits macrophage apoptosis and pro-inflammatory response through AKT-mediated regulation of β-catenin and FOXO-1. Cell Death Differ 2016; 23:1815-1826. [PMID: 27662364 DOI: 10.1038/cdd.2016.101] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/07/2016] [Revised: 07/18/2016] [Accepted: 08/17/2016] [Indexed: 12/25/2022] Open
Abstract
In order to establish infection, intra-macrophage parasite Leishmania donovani needs to inhibit host defense parameters like inflammatory cytokine production and apoptosis. In the present study, we demonstrate that the parasite achieves both by exploiting a single host regulator AKT for modulating its downstream transcription factors, β-catenin and FOXO-1. L. donovani-infected RAW264.7 and bone marrow-derived macrophages (BMDM) treated with AKT inhibitor or dominant negative AKT constructs showed decreased anti-inflammatory cytokine production and increased host cell apoptosis resulting in reduced parasite survival. Infection-induced activated AKT triggered phosphorylation-mediated deactivation of its downstream target, GSK-3β. Inactivated GSK-3β, in turn, could no longer sequester cytosolic β-catenin, an anti-apoptotic transcriptional regulator, as evidenced from its nuclear translocation during infection. Constitutively active GSK-3β-transfected L. donovani-infected cells mimicked the effects of AKT inhibition and siRNA-mediated silencing of β-catenin led to disruption of mitochondrial potential along with increased caspase-3 activity and IL-12 production leading to decreased parasite survival. In addition to activating anti-apoptotic β-catenin, phospho-AKT inhibits activation of FOXO-1, a pro-apoptotic transcriptional regulator. Nuclear retention of FOXO-1, inhibited during infection, was reversed when infected cells were transfected with dominant negative AKT constructs. Overexpression of FOXO-1 in infected macrophages not only documented increased apoptosis but promoted enhanced TLR4 expression and NF-κB activity along with an increase in IL-1β and decrease in IL-10 secretion. In vivo administration of AKT inhibitor significantly decreased liver and spleen parasite burden and switched cytokine balance in favor of host. In contrast, GSK-3β inhibitor did not result in any significant change in infectivity parameters. Collectively our findings revealed that L. donovani triggered AKT activation to regulate GSK-3β/β-catenin/FOXO-1 axis, thus ensuring inhibition of both host cell apoptosis and immune response essential for its intra-macrophage survival.
Collapse
Affiliation(s)
- Purnima Gupta
- Department of Biochemistry, University of Calcutta, Kolkata 700019, India
| | - Supriya Srivastav
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Shriya Saha
- Department of Biochemistry, University of Calcutta, Kolkata 700019, India
| | - Pijush K Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Anindita Ukil
- Department of Biochemistry, University of Calcutta, Kolkata 700019, India
| |
Collapse
|
36
|
Attenuated Leishmania induce pro-inflammatory mediators and influence leishmanicidal activity by p38 MAPK dependent phagosome maturation in Leishmania donovani co-infected macrophages. Sci Rep 2016; 6:22335. [PMID: 26928472 PMCID: PMC4772118 DOI: 10.1038/srep22335] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/11/2015] [Accepted: 02/08/2016] [Indexed: 12/21/2022] Open
Abstract
Promastigote form of Leishmania, an intracellular pathogen, delays phagosome maturation and resides inside macrophages. But till date limited study has been done to manipulate the phagosomal machinery of macrophages to restrict Leishmania growth. Attenuated Leishmania strain exposed RAW 264.7 cells showed a respiratory burst and enhanced production of pro-inflammatory mediators. The augmentation of pro-inflammatory activity is mostly attributed to p38 MAPK and p44/42 MAPK. In our study, these activated macrophages are found to induce phagosome maturation when infected with pathogenic Leishmania donovani. Increased co-localization of carboxyfluorescein succinimidyl ester labeled pathogenic L. donovani with Lysosome was found. Moreover, increased co-localization was observed between pathogenic L. donovani and late phagosomal markers viz. Rab7, Lysosomal Associated Membrane Protein 1, Cathepsin D, Rab9, and V-ATPase which indicate phagosome maturation. It was also observed that inhibition of V-type ATPase caused significant hindrance in attenuated Leishmania induced phagosome maturation. Finally, it was confirmed that p38 MAPK is the key player in acidification and maturation of phagosome in attenuated Leishmania strain pre-exposed macrophages. To our knowledge, this study for the first time reported an approach to induce phagosome maturation in L. donovani infected macrophages which could potentiate short-term prophylactic response in future.
Collapse
|
37
|
Soares-Silva M, Diniz FF, Gomes GN, Bahia D. The Mitogen-Activated Protein Kinase (MAPK) Pathway: Role in Immune Evasion by Trypanosomatids. Front Microbiol 2016; 7:183. [PMID: 26941717 PMCID: PMC4764696 DOI: 10.3389/fmicb.2016.00183] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/23/2015] [Accepted: 02/02/2016] [Indexed: 01/08/2023] Open
Abstract
Leishmania spp. and Trypanosoma cruzi are the causative agents of leishmaniasis and Chagas disease, respectively, two neglected tropical diseases that affect about 25 million people worldwide. These parasites belong to the family Trypanosomatidae, and are both obligate intracellular parasites that manipulate host signaling pathways and the innate immune system to establish infection. Mitogen-activated protein kinases (MAPKs) are serine and threonine protein kinases that are highly conserved in eukaryotes, and are involved in signal transduction pathways that modulate physiological and pathophysiological cell responses. This mini-review highlights existing knowledge concerning the mechanisms that Leishmania spp. and T. cruzi have evolved to target the host’s MAPK signaling pathways and highjack the immune response, and, in this manner, promote parasite maintenance in the host.
Collapse
Affiliation(s)
- Mercedes Soares-Silva
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Minas Gerais, Brazil
| | - Flavia F Diniz
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Minas Gerais, Brazil
| | - Gabriela N Gomes
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Minas Gerais, Brazil
| | - Diana Bahia
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas GeraisMinas Gerais, Brazil; Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São PauloSão Paulo, Brazil
| |
Collapse
|
38
|
Soares-Silva M, Diniz FF, Gomes GN, Bahia D. The Mitogen-Activated Protein Kinase (MAPK) Pathway: Role in Immune Evasion by Trypanosomatids. Front Microbiol 2016. [PMID: 26941717 DOI: 10.3389/fmicb.2016.00183/abstract] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 04/29/2023] Open
Abstract
Leishmania spp. and Trypanosoma cruzi are the causative agents of leishmaniasis and Chagas disease, respectively, two neglected tropical diseases that affect about 25 million people worldwide. These parasites belong to the family Trypanosomatidae, and are both obligate intracellular parasites that manipulate host signaling pathways and the innate immune system to establish infection. Mitogen-activated protein kinases (MAPKs) are serine and threonine protein kinases that are highly conserved in eukaryotes, and are involved in signal transduction pathways that modulate physiological and pathophysiological cell responses. This mini-review highlights existing knowledge concerning the mechanisms that Leishmania spp. and T. cruzi have evolved to target the host's MAPK signaling pathways and highjack the immune response, and, in this manner, promote parasite maintenance in the host.
Collapse
Affiliation(s)
- Mercedes Soares-Silva
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Minas Gerais, Brazil
| | - Flavia F Diniz
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Minas Gerais, Brazil
| | - Gabriela N Gomes
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais Minas Gerais, Brazil
| | - Diana Bahia
- Departamento de Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas GeraisMinas Gerais, Brazil; Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São PauloSão Paulo, Brazil
| |
Collapse
|
39
|
Ganguli P, Chowdhury S, Chowdhury S, Sarkar RR. Identification of Th1/Th2 regulatory switch to promote healing response during leishmaniasis: a computational approach. EURASIP JOURNAL ON BIOINFORMATICS & SYSTEMS BIOLOGY 2015; 2015:13. [PMID: 26660865 PMCID: PMC4666900 DOI: 10.1186/s13637-015-0032-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 08/04/2015] [Accepted: 11/18/2015] [Indexed: 11/10/2022]
Abstract
Leishmania devices its survival strategy by suppressing the host's immune functions. The antigen molecules produced by Leishmania interferes with the host's cell signaling cascades and consequently changes the protein expression pattern of the antigen-presenting cell (APC). This creates an environment suitable for the switching of the T-cell responses from a healing Th1 response to a non-healing Th2 response that is favorable for the continued survival of the parasite inside the host APC. Using a reconstructed signaling network of the intracellular and intercellular reactions between a Leishmania infected APC and T-cell, we propose a computational model to predict the inhibitory effect of the Leishmania infected APC on the T-cell and to identify the regulators of this Th1-/Th2-switching behavior as observed during Leishmania infection. In this work, we hypothesize that a complete removal of the parasite could only be achieved with a simultaneous up-regulation of the healing Th1 response and stimulation of nitric oxide (NO) production from the APCs, and downregulation of the non-healing Th2 response and thereby propose several unique combinations of protein molecules that could elicit this anti-Leishmania immune response. Our results indicate that TLR3 may play a positive role in eliciting NO synthesis, while TLR2 may be responsible for inhibiting an anti-Leishmania immune response. Also, TLR3 overexpression (in the APC), when combined with SHP2 inhibition (in the T cell), produces an anti-Leishmania response that is better than the conventional IFN-gamma or IL12 treatment. A similar anti-Leishmania response is also obtained in another combination where TLR3 (in APC) is overexpressed, and SHC and MKP (of T cell) are inhibited and activated, respectively. Through our study, we also observe that Leishmania infection may induce an upregulation of IFN-beta production from the APC that may lead to an upregulation of the RAP1 and SOCS3 proteins inside the T cell, the potential inhibitors of MAPK and JAK-STAT signaling pathways, respectively, via the TYK2-mediated pathway. This study not only enhances our knowledge in understanding the Th1/Th2 regulatory switch to promote healing response during leishmaniasis but also helps to identify novel combinations of proteins as potential immunomodulators.
Collapse
Affiliation(s)
- Piyali Ganguli
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune, Maharashtra 411008 India
| | - Saikat Chowdhury
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune, Maharashtra 411008 India ; Academy of Scientific & Innovative Research (AcSIR), CSIR-National Chemical Laboratory, Pune, India
| | - Shomeek Chowdhury
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune, Maharashtra 411008 India
| | - Ram Rup Sarkar
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune, Maharashtra 411008 India ; Academy of Scientific & Innovative Research (AcSIR), CSIR-National Chemical Laboratory, Pune, India
| |
Collapse
|
40
|
García-Hernández R, Gómez-Pérez V, Castanys S, Gamarro F. Fitness of Leishmania donovani parasites resistant to drug combinations. PLoS Negl Trop Dis 2015; 9:e0003704. [PMID: 25849149 PMCID: PMC4388546 DOI: 10.1371/journal.pntd.0003704] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/16/2015] [Accepted: 03/15/2015] [Indexed: 12/15/2022] Open
Abstract
Drug resistance represents one of the main problems for the use of chemotherapy to treat leishmaniasis. Additionally, it could provide some advantages to Leishmania parasites, such as a higher capacity to survive in stress conditions. In this work, in mixed populations of Leishmania donovani parasites, we have analyzed whether experimentally resistant lines to one or two combined anti-leishmanial drugs better support the stress conditions than a susceptible line expressing luciferase (Luc line). In the absence of stress, none of the Leishmania lines showed growth advantage relative to the other when mixed at a 1:1 parasite ratio. However, when promastigotes from resistant lines and the Luc line were mixed and exposed to different stresses, we observed that the resistant lines are more tolerant of different stress conditions: nutrient starvation and heat shock-pH stress. Further to this, we observed that intracellular amastigotes from resistant lines present a higher capacity to survive inside the macrophages than those of the control line. These results suggest that resistant parasites acquire an overall fitness increase and that resistance to drug combinations presents significant differences in their fitness capacity versus single-drug resistant parasites, particularly in intracellular amastigotes. These results contribute to the assessment of the possible impact of drug resistance on leishmaniasis control programs. Chemotherapy is currently the only treatment option for leishmaniasis, a neglected tropical disease produced by the protozoan parasite Leishmania. However, first-line drugs have different types of limitations including toxicity, price, efficacy and mainly emerging resistance. The WHO has recently recommended a combined therapy in order to extend the life expectancy of these compounds. The emergence and spread of Leishmania antimonial-resistant parasites have led to a high rate of antimonial failure in India and have raised questions about the selection and propagation risk of drug resistant parasites. The spread of drug-resistant parasites in the field probably depends on their transmission potential, which is influenced by, among other factors, the relative fitness of drug-resistant versus drug-susceptible parasites. In light of this, we have designed experimental studies to determine whether Leishmania donovani parasites resistant to single and combinations of anti-leishmanial drugs present any advantages in their ability to bear the different stress conditions versus a susceptible L. donovani line. Our results suggest that resistant parasites acquire an overall fitness increase and that resistance to drug combinations presents significant differences in their fitness capacity, particularly in intracellular amastigotes.
Collapse
Affiliation(s)
- Raquel García-Hernández
- Instituto de Parasitología y Biomedicina “López-Neyra”, IPBLN-CSIC, Parque Tecnológico de Ciencias de la Salud, Granada, Spain
| | - Verónica Gómez-Pérez
- Instituto de Parasitología y Biomedicina “López-Neyra”, IPBLN-CSIC, Parque Tecnológico de Ciencias de la Salud, Granada, Spain
| | - Santiago Castanys
- Instituto de Parasitología y Biomedicina “López-Neyra”, IPBLN-CSIC, Parque Tecnológico de Ciencias de la Salud, Granada, Spain
| | - Francisco Gamarro
- Instituto de Parasitología y Biomedicina “López-Neyra”, IPBLN-CSIC, Parque Tecnológico de Ciencias de la Salud, Granada, Spain
- * E-mail:
| |
Collapse
|
41
|
Chakraborty S, Srivastava A, Jha MK, Nair A, Pandey SP, Srivastava N, Kumari S, Singh S, Krishnasastry MV, Saha B. Inhibition of CD40-induced N-Ras activation reduces leishmania major infection. THE JOURNAL OF IMMUNOLOGY 2015; 194:3852-60. [PMID: 25786685 DOI: 10.4049/jimmunol.1401996] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 08/08/2014] [Accepted: 02/02/2015] [Indexed: 11/19/2022]
Abstract
Leishmania major is a parasite that resides and replicates in macrophages. We previously showed that the parasite enhanced CD40-induced Raf-MEK-ERK signaling but inhibited PI3K-MKK-p38MAPK signaling to proleishmanial effects. As Raf and PI3K have a Ras-binding domain but exert opposite effects on Leishmania infection, we examined whether Ras isoforms had differential roles in Leishmania infection. We observed that L. major enhanced N-Ras and H-Ras expression but inhibited K-Ras expression in macrophages. L. major infection enhanced N-Ras activity but inhibited H-Ras and K-Ras activity. TLR2 short hairpin RNA or anti-TLR2 or anti-lipophosphoglycan Abs reversed the L. major-altered N-Ras and K-Ras expressions. Pam3CSK4, a TLR2 ligand, enhanced N-Ras expression but reduced K-Ras expression, indicating TLR2-regulated Ras expression in L. major infection. Whereas N-Ras silencing reduced L. major infection, K-Ras and H-Ras silencing enhanced the infection both in macrophages in vitro and in C57BL/6 mice. BALB/c-derived macrophages transduced with lentivirally expressed N-Ras short hairpin RNA and pulsed with L. major-expressed MAPK10 enhanced MAPK10-specific Th1-type response. CD40-deficient mice primed with these macrophages had reduced L. major infection, accompanied by higher IFN-γ but less IL-4 production. As N-Ras is activated by Sos, a guanine nucleotide exchange factor, we modeled the N-Ras-Sos interaction and designed two peptides from their interface. Both the cell-permeable peptides reduced L. major infection in BALB/c mice but not in CD40-deficient mice. These data reveal the L. major-enhanced CD40-induced N-Ras activation as a novel immune evasion strategy and the potential for Ras isoform-targeted antileishmanial immunotherapy and immunoprophylaxis.
Collapse
Affiliation(s)
| | | | - Mukesh Kumar Jha
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Arathi Nair
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | | | - Neetu Srivastava
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Sangeeta Kumari
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Shailza Singh
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | | | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| |
Collapse
|
42
|
Chowdhury BP, Bandyopadhyay S, Das S, Majumder S, Jha MK, Majumdar SB, Saha B, Majumdar S. The host-protective effect of arabinosylated lipoarabinomannan against Leishmania donovani infection is associated with restoration of IFN-γ responsiveness. PLoS One 2015; 10:e0117247. [PMID: 25658110 PMCID: PMC4319725 DOI: 10.1371/journal.pone.0117247] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/11/2014] [Accepted: 12/22/2014] [Indexed: 12/20/2022] Open
Abstract
Visceral leishmaniasis (VL), which is endemic as a major infectious disease in the tropical and subtropical countries, is caused by a protozoan parasite Leishmania donovani. At present, restricted treatment options and lack of vaccines intensify the problem of controlling VL. Therefore, finding a novel immunoprophylactic or therapeutic principle is a pressing need. Here, we report that arabinosylated lipoarabinomannan (Ara-LAM), a TLR2-ligand isolated from Mycobacterium smegmatis, exhibits a strong immunomodulatory property that conferred protection against L. donovani infection. Although, Ara-LAM modulates TLR2 and MAPK signaling, it is not known whether Ara-LAM involves IFN-γ signaling for effective parasite clearance. Because, it is reported that IFN-γ signaling, a principle mediator of NO generation and macrophage and Tcell activation, is hampered during leishmanial pathogenesis. Ara-LAM increases IFN-γ receptor expression and potentiates IFN-γ receptor signaling through JAK-STAT pathway. Moreover, Ara-LAM reciprocally modulates IRF4 and IRF8 expression and reinstates anti-leishmanial Th1 response that eventuates in significantly reduced parasite load in spleen and liver of L. donovani-infected BALB/c mice. IFN-γRα silencing resulted in the suppression of these host-protective mechanisms affected by Ara-LAM. Thus, Ara-LAM-mediated restoration of IFN-γ responsiveness is a novel immuno-modulatory principle for protection against L. donovani susceptible host.
Collapse
Affiliation(s)
| | | | - Shibali Das
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Saikat Majumder
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Mukesh Kumar Jha
- Laboratory-V, National Centre for Cell Science (NCCS), Pune, Maharashtra, India
| | | | - Bhaskar Saha
- Laboratory-V, National Centre for Cell Science (NCCS), Pune, Maharashtra, India
| | - Subrata Majumdar
- Division of Molecular Medicine, Bose Institute, Kolkata, India
- * E-mail:
| |
Collapse
|
43
|
Marr AK, MacIsaac JL, Jiang R, Airo AM, Kobor MS, McMaster WR. Leishmania donovani infection causes distinct epigenetic DNA methylation changes in host macrophages. PLoS Pathog 2014; 10:e1004419. [PMID: 25299267 PMCID: PMC4192605 DOI: 10.1371/journal.ppat.1004419] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/28/2014] [Accepted: 08/23/2014] [Indexed: 12/15/2022] Open
Abstract
Infection of macrophages by the intracellular protozoan Leishmania leads to down-regulation of a number of macrophage innate host defense mechanisms, thereby allowing parasite survival and replication. The underlying molecular mechanisms involved remain largely unknown. In this study, we assessed epigenetic changes in macrophage DNA methylation in response to infection with L. donovani as a possible mechanism for Leishmania driven deactivation of host defense. We quantified and detected genome-wide changes of cytosine methylation status in the macrophage genome resulting from L. donovani infection. A high confidence set of 443 CpG sites was identified with changes in methylation that correlated with live L. donovani infection. These epigenetic changes affected genes that play a critical role in host defense such as the JAK/STAT signaling pathway and the MAPK signaling pathway. These results provide strong support for a new paradigm in host-pathogen responses, where upon infection the pathogen induces epigenetic changes in the host cell genome resulting in downregulation of innate immunity thereby enabling pathogen survival and replication. We therefore propose a model whereby Leishmania induced epigenetic changes result in permanent down regulation of host defense mechanisms to protect intracellular replication and survival of parasitic cells. The L. donovani parasite causes visceral leishmaniasis, a tropical, neglected disease with an estimated number of 500,000 cases worldwide. Current drug treatments have toxic side effects, lead to drug resistance, and an effective vaccine is not available. The parasite has a complex life cycle residing within different host environments including the gut of a sand fly and immune cells of the mammalian host. Alteration of host cell gene expression including signaling pathways has been shown to be a major strategy to evade host cell immune response and thus enables the Leishmania parasite to survive, replicate and persist in its host cells. Recently it was demonstrated that intracellular pathogens such as viruses and bacteria are able to manipulate epigenetic processes, thereby perhaps facilitating their intracellular survival. Using an unbiased genome-wide DNA methylation approach, we demonstrate here that an intracellular parasite can alter host cell DNA methylation patterns resulting in altered gene expression possibly to establish disease. Thus DNA methylation changes in host cells upon infection might be a common strategy among intracellular pathogens for their uncontrolled replication and dissemination.
Collapse
Affiliation(s)
- Alexandra K. Marr
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Julia L. MacIsaac
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Ruiwei Jiang
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Adriana M. Airo
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Michael S. Kobor
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- Human Early Learning Partnership, School of Population and Public Health, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- * E-mail: (MSK); (WRM)
| | - W. Robert McMaster
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- * E-mail: (MSK); (WRM)
| |
Collapse
|
44
|
Pratap Singh K, Zaidi A, Anwar S, Bimal S, Das P, Ali V. Reactive oxygen species regulates expression of iron-sulfur cluster assembly protein IscS of Leishmania donovani. Free Radic Biol Med 2014; 75:195-209. [PMID: 25062827 DOI: 10.1016/j.freeradbiomed.2014.07.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/08/2014] [Revised: 07/10/2014] [Accepted: 07/14/2014] [Indexed: 01/18/2023]
Abstract
The cysteine desulfurase, IscS, is a highly conserved and essential component of the mitochondrial iron-sulfur cluster (ISC) system that serves as a sulfur donor for Fe-S clusters biogenesis. Fe-S clusters are versatile and labile cofactors of proteins that orchestrate a wide array of essential metabolic processes, such as energy generation and ribosome biogenesis. However, no information regarding the role of IscS or its regulation is available in Leishmania, an evolving pathogen model with rapidly developing drug resistance. In this study, we characterized LdIscS to investigate the ISC system in AmpB-sensitive vs resistant isolates of L. donovani and to understand its regulation. We observed an upregulated Fe-S protein activity in AmpB-resistant isolates but, in contrast to our expectations, LdIscS expression was upregulated in the sensitive strain. However, further investigations showed that LdIscS expression is positively correlated with ROS level and negatively correlated with Fe-S protein activity, independent of strain sensitivity. Thus, our results suggested that LdIscS expression is regulated by ROS level with Fe-S clusters/proteins acting as ROS sensors. Moreover, the direct evidence of a mechanism, in support of our results, is provided by dose-dependent induction of LdIscS-GFP as well as endogenous LdIscS in L. donovani promastigotes by three different ROS inducers: H2O2, menadione, and Amphotericin B. We postulate that LdIscS is upregulated for de novo synthesis or repair of ROS damaged Fe-S clusters. Our results reveal a novel mechanism for regulation of IscS expression that may help parasite survival under oxidative stress conditions encountered during infection of macrophages and suggest a cross talk between two seemingly unrelated metabolic pathways, the ISC system and redox metabolism in L. donovani.
Collapse
Affiliation(s)
- Krishn Pratap Singh
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, India 800007
| | - Amir Zaidi
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, India 800007
| | - Shadab Anwar
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, India 800007
| | - Sanjeev Bimal
- Department of Immunology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, India 800007
| | - Pradeep Das
- Department of Molecular Biology, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, India 800007
| | - Vahab Ali
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna, India 800007.
| |
Collapse
|
45
|
Dan-Goor M, Nasereddin A, Jaber H, Jaffe CL. Identification of a secreted casein kinase 1 in Leishmania donovani: effect of protein over expression on parasite growth and virulence. PLoS One 2013; 8:e79287. [PMID: 24260187 PMCID: PMC3829951 DOI: 10.1371/journal.pone.0079287] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/24/2013] [Accepted: 09/25/2013] [Indexed: 12/14/2022] Open
Abstract
Casein kinase 1 (CK1) plays an important role in eukaryotic signaling pathways, and their substrates include key regulatory proteins involved in cell differentiation, proliferation and chromosome segregation. The Leishmania genome encodes six potential CK1 isoforms, of which five have orthologs in other trypanosomatidae. Leishmania donovani CK1 isoform 4 (Ldck1.4, orthologous to LmjF27.1780) is unique to Leishmania and contains a putative secretion signal peptide. The full-length gene and three shorter constructs were cloned and expressed in E. coli as His-tag proteins. Only the full-length 62.3 kDa protein showed protein kinase activity indicating that the N-terminal and C-terminal domains are essential for protein activity. LdCK1.4-FLAG was stably over expressed in L. donovani, and shown by immunofluorescence to be localized primarily in the cytosol. Western blotting using anti-FLAG and anti-CK1.4 antibodies showed that this CK1 isoform is expressed and secreted by promastigotes. Over expression of LdCK1.4 had a significant effect on promastigote growth in culture with these parasites growing to higher cell densities than the control parasites (wild-type or Ld:luciferase, P<0.001). Analysis by flow cytometry showed a higher percentage, ∼4-5-fold, of virulent metacyclic promastigotes on day 3 among the LdCK1.4 parasites. Finally, parasites over expressing LdCK1.4 gave significantly higher infections of mouse peritoneal macrophages compared to wild-type parasites, 28.6% versus 6.3%, respectively (p = 0.0005). These results suggest that LdCK1.4 plays an important role in parasite survival and virulence. Further studies are needed to validate CK1.4 as a therapeutic target in Leishmania.
Collapse
Affiliation(s)
- Mary Dan-Goor
- Department of Microbiology and Molecular Genetics, The Kuvin Center for the Study of Infectious and Tropical Diseases, National Center for Leishmaniasis, IMRIC, Hebrew University–Hadassah Medical School, Jerusalem, Israel
| | - Abedelmajeed Nasereddin
- Department of Microbiology and Molecular Genetics, The Kuvin Center for the Study of Infectious and Tropical Diseases, National Center for Leishmaniasis, IMRIC, Hebrew University–Hadassah Medical School, Jerusalem, Israel
| | - Hanan Jaber
- Department of Microbiology and Molecular Genetics, The Kuvin Center for the Study of Infectious and Tropical Diseases, National Center for Leishmaniasis, IMRIC, Hebrew University–Hadassah Medical School, Jerusalem, Israel
| | - Charles L. Jaffe
- Department of Microbiology and Molecular Genetics, The Kuvin Center for the Study of Infectious and Tropical Diseases, National Center for Leishmaniasis, IMRIC, Hebrew University–Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
46
|
Lemaire J, Mkannez G, Guerfali FZ, Gustin C, Attia H, Sghaier RM, Dellagi K, Laouini D, Renard P. MicroRNA expression profile in human macrophages in response to Leishmania major infection. PLoS Negl Trop Dis 2013; 7:e2478. [PMID: 24098824 PMCID: PMC3789763 DOI: 10.1371/journal.pntd.0002478] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/07/2013] [Accepted: 08/30/2013] [Indexed: 12/31/2022] Open
Abstract
Background Leishmania (L.) are intracellular protozoan parasites able to survive and replicate in the hostile phagolysosomal environment of infected macrophages. They cause leishmaniasis, a heterogeneous group of worldwide-distributed affections, representing a paradigm of neglected diseases that are mainly embedded in impoverished populations. To establish successful infection and ensure their own survival, Leishmania have developed sophisticated strategies to subvert the host macrophage responses. Despite a wealth of gained crucial information, these strategies still remain poorly understood. MicroRNAs (miRNAs), an evolutionarily conserved class of endogenous 22-nucleotide non-coding RNAs, are described to participate in the regulation of almost every cellular process investigated so far. They regulate the expression of target genes both at the levels of mRNA stability and translation; changes in their expression have a profound effect on their target transcripts. Methodology/Principal Findings We report in this study a comprehensive analysis of miRNA expression profiles in L. major-infected human primary macrophages of three healthy donors assessed at different time-points post-infection (three to 24 h). We show that expression of 64 out of 365 analyzed miRNAs was consistently deregulated upon infection with the same trends in all donors. Among these, several are known to be induced by TLR-dependent responses. GO enrichment analysis of experimentally validated miRNA-targeted genes revealed that several pathways and molecular functions were disturbed upon parasite infection. Finally, following parasite infection, miR-210 abundance was enhanced in HIF-1α-dependent manner, though it did not contribute to inhibiting anti-apoptotic pathways through pro-apoptotic caspase-3 regulation. Conclusions/Significance Our data suggest that alteration in miRNA levels likely plays an important role in regulating macrophage functions following L. major infection. These results could contribute to better understanding of the dynamics of gene expression in host cells during leishmaniasis. Leishmania parasites belong to different species, each one characterized by specific vectors and reservoirs, and causes cutaneous or visceral disease(s) of variable clinical presentation and severity. In its mammalian host, the parasite is an obligate intracellular pathogen infecting the monocyte/macrophage lineage. Leishmania have developed ambiguous relationships with macrophages. Indeed, these cells are the shelter of invading parasites, where they will grow and eventually will reside in a silent state for life. But macrophages are also the cells that participate, through the induction of several pro-inflammatory mediators and antigen presentation, to shape the host immune response and ultimately kill the invader. To subvert these anti-parasite responses, Leishmania manipulate the host machinery for their own differentiation and survival. We aimed to evaluate the impact of L. major (the causative agent of zoonotic cutaneous leishmaniasis) infection on deregulation of non-coding miRNAs, a class of important regulators of gene expression. Our results revealed the implication of several miRNAs on macrophage fate upon parasite infection through regulation of different pathways, including cell death. Our findings provided a new insight for understanding mechanisms governing this miRNA deregulation by parasite infection and will help to provide clues for the development of control strategies for this disease.
Collapse
Affiliation(s)
- Julien Lemaire
- Laboratory of Biochemistry and Cellular Biology (URBC), NARILIS-University of Namur, Namur, Belgium
| | - Ghada Mkannez
- Institut Pasteur de Tunis, LR11IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
| | - Fatma Z. Guerfali
- Institut Pasteur de Tunis, LR11IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
| | - Cindy Gustin
- Laboratory of Biochemistry and Cellular Biology (URBC), NARILIS-University of Namur, Namur, Belgium
| | - Hanène Attia
- Institut Pasteur de Tunis, LR11IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
| | - Rabiaa M. Sghaier
- Institut Pasteur de Tunis, LR11IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
| | | | - Koussay Dellagi
- Institut Pasteur de Tunis, LR11IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
- Institut de Recherche pour le Développement (IRD) et Centre de Recherche et de Veille sur les Maladies Emergentes dans l'Océan Indien (CRVOI), Sainte Clotilde, Reunion Island, France
| | - Dhafer Laouini
- Institut Pasteur de Tunis, LR11IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
- * E-mail: , (DL); (PR)
| | - Patricia Renard
- Laboratory of Biochemistry and Cellular Biology (URBC), NARILIS-University of Namur, Namur, Belgium
- * E-mail: , (DL); (PR)
| |
Collapse
|
47
|
Kumar V, Gour JK, Singh N, Bajpai S, Singh RK. Leishmania donovani-specific 25- and 28-kDa urinary proteins activate macrophage effector functions, lymphocyte proliferation and Th1 cytokines production. Parasitol Res 2013; 112:1427-35. [PMID: 23334693 DOI: 10.1007/s00436-013-3272-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/01/2012] [Accepted: 01/03/2013] [Indexed: 10/27/2022]
Abstract
Growing incidence of drug resistance against leishmaniasis in endemic areas and limited drug options necessitates the need for a vaccine. Notwithstanding significant leishmanial research in the past decades, a vaccine candidate is far from reality. In this study, we report the potential of two urinary leishmanial proteins to induce macrophage effector functions, inflammatory cytokines production and human lymphocytes proliferation. A total four proteins of molecular mass 25, 28, 54 and 60 kDa were identified in human urine samples. The 25 and 28 kDa proteins significantly induced NADPH oxidase (p<0.001), superoxide dismutase (p<0.001) and inducible nitric oxide synthase (p<0.001) activities in stimulated RAW264.7 macrophages. The release of nitric oxide, tumor necrosis factor-alpha and interleukin (IL)-12 was also significantly (p<0.001) higher in 25 and 28 kDa activated macrophages as compared with cells activated with other two proteins. These two proteins also induced significant (p<0.001) proliferation and release of IFN-γ and IL-12 in human peripheral blood mononuclear cells.
Collapse
Affiliation(s)
- Vinod Kumar
- Molecular Immunology Laboratory, Department of Biochemistry, Faculty of Science, Banaras Hindu University, Varanasi, 221 005, India
| | | | | | | | | |
Collapse
|
48
|
Infection by Leishmania amazonensis in mice: a potential model for chronic hypoxia. Acta Histochem 2012; 114:797-804. [PMID: 22360823 DOI: 10.1016/j.acthis.2012.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/03/2011] [Revised: 01/28/2012] [Accepted: 01/30/2012] [Indexed: 11/20/2022]
Abstract
Hypoxia is a common feature of injured and infected tissues. Hypoxia inducible factors 1α and 2α (HIF-1α, HIF-2α) are heterodimeric transcription factors mediating the cellular responses to hypoxia and also the vascular endothelial growth factor (VEGF). VEGF is a cytokine which can be induced by hypoxia, whose pathogenic mechanisms are still unclear and which is the subject of debate. Murine cutaneous lesions during Leishmania amazonensis parasite infection are chronic, although they are small and self-controlled in C57BL/6 mice and severe in BALB/c mice. In the present study we examined the presence of hypoxia, HIF-1α, HIF-2α and VEGF during the course of infection in both mouse strains. Hypoxia was detected in lesions from BALB/c mice by pimonidazole marking, which occurred earlier than in lesions from C57Bl/6 mice. The lesions in the BALB/c mice showed HIF-1α and HIF-2α expression in the cytoplasm of macrophages and failed to promote any VEGF expression, while lesions in the C57BL/6 mice showed HIF-2α nuclear accumulation and subsequent VEGF expression. In conclusion, the animal models of leishmaniasis demonstrated a diversity of patterns of expression, cell localization and activity of the main transducers of hypoxia and may be useful models for studying the pathogenic mechanisms of HIF-1α and HIF-2α during chronic hypoxic diseases.
Collapse
|
49
|
Soong L, Henard CA, Melby PC. Immunopathogenesis of non-healing American cutaneous leishmaniasis and progressive visceral leishmaniasis. Semin Immunopathol 2012; 34:735-51. [PMID: 23053396 PMCID: PMC4111229 DOI: 10.1007/s00281-012-0350-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/02/2012] [Accepted: 09/21/2012] [Indexed: 12/21/2022]
Abstract
The outcomes of Leishmania infection are determined by host immune and nutrition status, parasite species, and co-infection with other pathogens. While subclinical infection and self-healing cutaneous leishmaniasis (CL) are common, uncontrolled parasite replication can lead to non-healing local lesions or visceral leishmaniasis (VL). It is known that infection control requires Th1-differentiation cytokines (IL-12, IL-18, and IL-27) and Th1 cell and macrophage activation. However, there is no generalized consensus for the mechanisms of host susceptibility. The recent studies on regulatory T cells and IL-17-producing cells help explain the effector T cell responses that occur independently of the known Th1/Th2 cell signaling pathways. This review focuses on the immunopathogenesis of non-healing American CL and progressive VL. We summarize recent evidence from human and animal studies that reveals the mechanisms of dysregulated, hyper-responses to Leishmania braziliensis, as well as the presence of disease-promoting or the absence of protective responses to Leishmania amazonensis and Leishmania donovani. We highlight immune-mediated parasite growth and immunopathogenesis, with an emphasis on the putative roles of IL-17 and its related cytokines as well as arginase. A better understanding of the quality and regulation of innate immunity and T cell responses triggered by Leishmania will aid in the rational control of pathology and the infection.
Collapse
Affiliation(s)
- Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555-1070, USA.
| | | | | |
Collapse
|
50
|
Vanaerschot M, Decuypere S, Berg M, Roy S, Dujardin JC. Drug-resistant microorganisms with a higher fitness--can medicines boost pathogens? Crit Rev Microbiol 2012; 39:384-94. [PMID: 22950457 DOI: 10.3109/1040841x.2012.716818] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/13/2022]
Abstract
Drug-resistant microorganisms (DRMs) are generally thought to suffer from a fitness cost associated with their drug-resistant trait, inflicting them a disadvantage when the drug pressure reduces. However, Leishmania resistant to pentavalent antimonies shows traits of a higher fitness compared to its sensitive counterparts. This is likely due the combination of an intracellular pathogen and a drug that targets the parasite's general defense mechanisms while at the same time stimulating the host's immune system, resulting in a DRM that is better adapted to withstand the host's immune response. This review aims to highlight how this fitter DRM has emerged and how it might affect the control of leishmaniasis. However, this unprecedented example of fitter antimony-resistant Leishmania donovani is also of significance for the control of other microorganisms, warranting more caution when applying or designing drugs that attack their general defense mechanisms or interact with the host's immune system.
Collapse
Affiliation(s)
- Manu Vanaerschot
- Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp , Antwerpen , Belgium
| | | | | | | | | |
Collapse
|