1
|
Sanku G, Ricciardi A, Redekar NR, Schaughency P, Lack J, Gazzinelli-Guimaraes PH, Nutman TB. Brugia malayi filarial helminth-derived extracellular vesicles suppress antigen presenting cell function and antigen-specific CD4+ T cell responses. Front Immunol 2024; 15:1436818. [PMID: 39434874 PMCID: PMC11491353 DOI: 10.3389/fimmu.2024.1436818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/08/2024] [Indexed: 10/23/2024] Open
Abstract
Introduction Live microfilariae (mf) and mf-derived extracellular vesicles (EVs) have been shown to modulate human antigen presenting cell (APC) function, most notably by suppressing the induction of IL-12 (and other pro-inflammatory cytokines) following activation with LPS and interferon-y. Methods To explore further how EVs alter human APC function, we studied the effect of mf and EVs on human elutriated monocyte-derived dendritic cells (DC) following exposure to Mf, mf-derived excretory/secretory (E/S) products, E/S depleted of EVs through ultracentrifugation and purified EVs. After demonstrating that the measurable responses induced by live mf could be recapitulated by EVs and EV-containing E/S, we next performed RNAseq analysis of human DC following exposure to live mf, EVs, E/S, or EV-depleted E/S. Results In our analyses of the data for the DC, using a false discovery rate (FDR)<0.05, EV-exposed DC had induced the expression of 212 differentially expressed genes (DEGs) when compared to unexposed DC and 157 when compared to E/S-depleted EVs. These genes were enriched in GO biological processes associated with neutrophil degranulation and 15 DEGs associated with KEGG Lysosome pathways. IPA analysis point to immune dysregulation. We next aimed to understand the intracellular processes altered by EVs and the effect these have on effector T cells. When SARS CoV-2 Membrane-specific CD4+ TCLs were assessed following EV conditioning of autologous DC and activation with the SARS CoV-2-Membrane peptide pool, we found conditioning reduced the frequency of SARS CoV-2 Membrane-specific CD3+ CD4+ CD154+ cells (p=.015). Similarly, EV-conditioning of SARS CoV-2 Membrane-specific CD3+ CD4+ cells induced fewer cell capable of producing IFN-γ (p=.045). Discussion Taken together, our data suggest a modulatory role of EVs on APC function that likely leads to defects in T cell effector function.
Collapse
Affiliation(s)
- Gayatri Sanku
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Alessandra Ricciardi
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Neelam R. Redekar
- Integrated Data Science Section (IDSS), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Paul Schaughency
- Integrated Data Science Section (IDSS), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Justin Lack
- Integrated Data Science Section (IDSS), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Pedro H. Gazzinelli-Guimaraes
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Thomas B. Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
2
|
Wang H, Li Y, Yu Q, Wang M, Ainiwaer A, Tang N, Zheng X, Duolikun A, Deng B, Li J, Shen Y, Zhang C. Immunological Characteristics of Hepatic Dendritic Cells in Patients and Mouse Model with Liver Echinococcus multilocularis Infection. Trop Med Infect Dis 2024; 9:95. [PMID: 38787028 PMCID: PMC11125766 DOI: 10.3390/tropicalmed9050095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
The cestode Echinococcus multilocularis, which mainly dwells in the liver, leads to a serious parasitic liver disease called alveolar echinococcosis (AE). Despite the increased attention drawn to the immunosuppressive microenvironment formed by hepatic AE tissue, the immunological characteristics of hepatic dendritic cells (DCs) in the AE liver microenvironment have not been fully elucidated. Here, we profiled the immunophenotypic characteristics of hepatic DC subsets in both clinical AE patients and a mouse model. Single-cell RNA sequencing (scRNA-Seq) analysis of four AE patient specimens revealed that greater DC numbers were present within perilesional liver tissues and that the distributions of cDC and pDC subsets in the liver and periphery were different. cDCs highly expressed the costimulatory molecule CD86, the immune checkpoint molecule CD244, LAG3, CTLA4, and the checkpoint ligand CD48, while pDCs expressed these genes at low frequencies. Flow cytometric analysis of hepatic DC subsets in an E. multilocularis infection mouse model demonstrated that the number of cDCs significantly increased after parasite infection, and a tolerogenic phenotype characterized by a decrease in CD40 and CD80 expression levels was observed at an early stage, whereas an activated phenotype characterized by an increase in CD86 expression levels was observed at a late stage. Moreover, the expression profiles of major immune checkpoint molecules (CD244 and LAG3) and ligands (CD48) on hepatic DC subsets in a mouse model exhibited the same pattern as those in AE patients. Notably, the cDC and pDC subsets in the E. multilocularis infection group exhibited higher expression levels of PD-L1 and CD155 than those in the control group, suggesting the potential of these subsets to impair T cell function. These findings may provide valuable information for investigating the role of hepatic DC subsets in the AE microenvironment and guiding DC targeting treatments for AE.
Collapse
Affiliation(s)
- Hui Wang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
- Basic Medical College, Xinjiang Medical University, Urumqi 830011, China
| | - Yinshi Li
- Basic Medical College, Xinjiang Medical University, Urumqi 830011, China
| | - Qian Yu
- Basic Medical College, Xinjiang Medical University, Urumqi 830011, China
| | - Mingkun Wang
- Basic Medical College, Xinjiang Medical University, Urumqi 830011, China
| | - Abidan Ainiwaer
- Basic Medical College, Xinjiang Medical University, Urumqi 830011, China
| | - Na Tang
- Basic Medical College, Xinjiang Medical University, Urumqi 830011, China
| | - Xuran Zheng
- Basic Medical College, Xinjiang Medical University, Urumqi 830011, China
| | - Adilai Duolikun
- Basic Medical College, Xinjiang Medical University, Urumqi 830011, China
| | - Bingqing Deng
- Basic Medical College, Xinjiang Medical University, Urumqi 830011, China
| | - Jing Li
- Basic Medical College, Xinjiang Medical University, Urumqi 830011, China
| | - Yujuan Shen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology (National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention), World Health Organization Collaborating Centre for Tropical Disease, Shanghai 200025, China
| | - Chuanshan Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
- Basic Medical College, Xinjiang Medical University, Urumqi 830011, China
| |
Collapse
|
3
|
Bowman C, Rockson SG. The Role of Inflammation in Lymphedema: A Narrative Review of Pathogenesis and Opportunities for Therapeutic Intervention. Int J Mol Sci 2024; 25:3907. [PMID: 38612716 PMCID: PMC11011271 DOI: 10.3390/ijms25073907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Lymphedema is a chronic and progressive disease of the lymphatic system characterized by inflammation, increased adipose deposition, and tissue fibrosis. Despite early hypotheses identifying lymphedema as a disease of mechanical lymphatic disruption alone, the progressive inflammatory nature underlying this condition is now well-established. In this review, we provide an overview of the various inflammatory mechanisms that characterize lymphedema development and progression. These mechanisms contribute to the acute and chronic phases of lymphedema, which manifest clinically as inflammation, fibrosis, and adiposity. Furthermore, we highlight the interplay between current therapeutic modalities and the underlying inflammatory microenvironment, as well as opportunities for future therapeutic development.
Collapse
Affiliation(s)
- Catharine Bowman
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA;
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Stanley G. Rockson
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA;
| |
Collapse
|
4
|
The Domestic Dog as a Laboratory Host for Brugia malayi. Pathogens 2022; 11:pathogens11101073. [PMID: 36297129 PMCID: PMC9607614 DOI: 10.3390/pathogens11101073] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/07/2022] [Accepted: 09/19/2022] [Indexed: 11/21/2022] Open
Abstract
Of the three nematodes responsible for lymphatic filariasis in humans, only Brugia malayi is actively maintained in research settings owing to its viability in small animal hosts, principal among which is the domestic cat. While the microfilaremic feline host is necessary for propagation of parasites on any significant scale, this system is plagued by a number of challenges not as pronounced in canine filarial models. For this reason, we investigated the capacity in which dogs may serve as competent laboratory hosts for B. malayi. We infected a total of 20 dogs by subcutaneous injection of 500 B. malayi third-stage larvae (L3) in either a single (n = 10) or repeated infection events (125 L3 per week for four weeks; n = 10). Within each group, half of the individuals were injected in the inguinal region and half in the dorsum of the hind paw. To track the course of microfilaremia in this host, blood samples were examined by microscopy biweekly for two years following infection. Additionally, to identify cellular responses with potential value as predictors of patency, we measured peripheral blood leukocyte counts for the first year of infection. A total of 10 of 20 dogs developed detectable microfilaremia. Peak microfilaria density varied but attained levels useful for parasite propagation (median = 1933 mL−1; range: 33–9950 mL−1). Nine of these dogs remained patent at 104 weeks. A two-way ANOVA revealed no significant differences between infection groups in lifetime microfilaria production (p = 0.42), nor did regression analysis reveal any likely predictive relationships to leukocyte values. The results of this study demonstrate the competence of the dog as a host for B. malayi and its potential to serve in the laboratory role currently provided by the cat, while also clarifying the potential for zoonosis in filariasis-endemic regions.
Collapse
|
5
|
Ricciardi A, Hassan SA, Kamenyeva O, Bennuru S, Andersen J, Nutman TB. A filarial parasite-encoded human IL-10 receptor antagonist reveals a novel strategy to modulate host responses. PNAS NEXUS 2022; 1:pgac184. [PMID: 36246151 PMCID: PMC9552326 DOI: 10.1093/pnasnexus/pgac184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/31/2022] [Indexed: 02/05/2023]
Abstract
Interleukin (IL)-10 is the primary cytokine driving the modulation of the host response in filarial infections. We performed binding assays with Brugia malayi antigen extracts and human IL-10R1. Bm5539 was the top-binding hit. We identified a short sequence, termed truncated Bm5339, that has structural similarities to the human IL-10 functional dimer. Sequence comparisons revealed that other filarial parasites possess Bm5539 orthologues. Using recombinant Bm5539 in a modified Luciferase Immunoprecipitation System assay, we confirmed that both the truncated and full-length forms of the protein can bind to human IL-10R1. Truncated Bm5539 could inhibit human IL-10-driven phosphorylation of STAT3, thereby demonstrating that Bm5539 acts as an IL-10 antagonist, most likely through competitive binding to the receptor. We provide a structural basis for these observations using computational modeling and simulations. This parasite-encoded cytokine receptor antagonist provides an additional lens through which parasite-induced modulation of the host immune response can be examined.
Collapse
Affiliation(s)
- Alessandra Ricciardi
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - Sergio A Hassan
- Bioinformatics and Computational Biosciences Branch, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - Olena Kamenyeva
- Research Technology Branch, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - Sasisekhar Bennuru
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| | - John Andersen
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, 12735 Twinbrook Parkway, Rockville, MD 20852, USA
| | | |
Collapse
|
6
|
Bhoj P, Togre N, Khatri V, Goswami K. Harnessing Immune Evasion Strategy of Lymphatic Filariae: A Therapeutic Approach against Inflammatory and Infective Pathology. Vaccines (Basel) 2022; 10:vaccines10081235. [PMID: 36016123 PMCID: PMC9415972 DOI: 10.3390/vaccines10081235] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 02/04/2023] Open
Abstract
Human lymphatic filariae have evolved numerous immune evasion strategies to secure their long-term survival in a host. These strategies include regulation of pattern recognition receptors, mimicry with host glycans and immune molecules, manipulation of innate and adaptive immune cells, induction of apoptosis in effector immune cells, and neutralization of free radicals. This creates an anti-inflammatory and immunoregulatory milieu in the host: a modified Th2 immune response. Therefore, targeting filarial immunomodulators and manipulating the filariae-driven immune system against the filariae can be a potential therapeutic and prophylactic strategy. Filariae-derived immunosuppression can also be exploited to treat other inflammatory diseases and immunopathologic states of parasitic diseases, such as cerebral malaria, and to prevent leishmaniasis. This paper reviews immunomodulatory mechanisms acquired by these filariae for their own survival and their potential application in the development of novel therapeutic approaches against parasitic and inflammatory diseases. Insight into the intricate network of host immune-parasite interactions would aid in the development of effective immune-therapeutic options for both infectious and immune-pathological diseases.
Collapse
Affiliation(s)
| | - Namdev Togre
- Department of Biological Sciences, University of Texas, El Paso, TX 79968, USA
- Correspondence: (N.T.); (K.G.)
| | | | - Kalyan Goswami
- All India Institute of Medical Sciences, Saguna, Kalyani 741245, India
- Correspondence: (N.T.); (K.G.)
| |
Collapse
|
7
|
Joshi P, Mishra PKK. Functional Diversity of the Excretory/Secretory Proteins of Nematode Parasites. Acta Parasitol 2022; 67:619-627. [PMID: 35113339 DOI: 10.1007/s11686-022-00523-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 01/18/2022] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Parasites release a wide array of protein as excretory and secretory products (ESPs). Irrespective of their mode of propagation, ESPs are found to be secreted or excreted by both naturally occurring and laboratory-cultivated parasites. Mass spectrometry-based approaches have been extensively used to identify and characterize the ESP constituents. ESPs are involved in various cellular activities such as immune modulation, proteolysis, inhibition of proteases and protection of cells against oxidants. Specifically, their role in host immune evasion by down-regulation of pro-inflammatory cytokines and up-regulation of anti-inflammatory cytokines attracts scientific attention. A thorough investigation of functional diversity of ESPs may be helpful in planning control strategies against many parasites. METHODS This review focuses on diversity of ES proteins, various approaches to identify them and discusses about the biochemical and functional aspects of such proteins. RESULTS The diverse array of proteins secreted or excreted (a, GST-1, acetylcholinesterase, GAPDH) by the parasites are also described emphasizing their role in cellular physiology. CONCLUSION Finally, it concludes by citing some of these proteins as potential therapeutic agents against helminth challenge.
Collapse
Affiliation(s)
- Paritosh Joshi
- Division of Biochemistry, ICAR-Indian Veterinary Research Institute, Izatnagar, U.P., 243122, India.
- Panchvati, Bijroli, Bhimtal, Uttarakhand, India.
| | - Prasanta Kumar K Mishra
- Division of Biochemistry, ICAR-Indian Veterinary Research Institute, Izatnagar, U.P., 243122, India.
- Unit of Veterinary Clinical Complex, Faculty of Veterinary and Animal Sciences, RGSC, Banaras Hindu University, Mirzapur, U.P., India.
| |
Collapse
|
8
|
Kraemer L, McKay DM, Russo RC, Fujiwara RT. Chemokines and chemokine receptors: insights from human disease and experimental models of helminthiasis. Cytokine Growth Factor Rev 2022; 66:38-52. [DOI: 10.1016/j.cytogfr.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 11/03/2022]
|
9
|
Bąska P, Norbury LJ. The Role of Nuclear Factor Kappa B (NF-κB) in the Immune Response against Parasites. Pathogens 2022; 11:pathogens11030310. [PMID: 35335634 PMCID: PMC8950322 DOI: 10.3390/pathogens11030310] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/17/2022] [Accepted: 02/25/2022] [Indexed: 12/28/2022] Open
Abstract
The immune system consists of various cells, organs, and processes that interact in a sophisticated manner to defend against pathogens. Upon initial exposure to an invader, nonspecific mechanisms are raised through the activation of macrophages, monocytes, basophils, mast cells, eosinophils, innate lymphoid cells, or natural killer cells. During the course of an infection, more specific responses develop (adaptive immune responses) whose hallmarks include the expansion of B and T cells that specifically recognize foreign antigens. Cell to cell communication takes place through physical interactions as well as through the release of mediators (cytokines, chemokines) that modify cell activity and control and regulate the immune response. One regulator of cell states is the transcription factor Nuclear Factor kappa B (NF-κB) which mediates responses to various stimuli and is involved in a variety of processes (cell cycle, development, apoptosis, carcinogenesis, innate and adaptive immune responses). It consists of two protein classes with NF-κB1 (p105/50) and NF-κB2 (p100/52) belonging to class I, and RelA (p65), RelB and c-Rel belonging to class II. The active transcription factor consists of a dimer, usually comprised of both class I and class II proteins conjugated to Inhibitor of κB (IκB). Through various stimuli, IκB is phosphorylated and detached, allowing dimer migration to the nucleus and binding of DNA. NF-κB is crucial in regulating the immune response and maintaining a balance between suppression, effective response, and immunopathologies. Parasites are a diverse group of organisms comprised of three major groups: protozoa, helminths, and ectoparasites. Each group induces distinct effector immune mechanisms and is susceptible to different types of immune responses (Th1, Th2, Th17). This review describes the role of NF-κB and its activity during parasite infections and its contribution to inducing protective responses or immunopathologies.
Collapse
Affiliation(s)
- Piotr Bąska
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, 02-786 Warsaw, Poland
- Correspondence:
| | - Luke J. Norbury
- Department of Biosciences and Food Technology, School of Science, STEM College, RMIT University, Bundoora, VIC 3083, Australia;
| |
Collapse
|
10
|
Chulanetra M, Chaicumpa W. Revisiting the Mechanisms of Immune Evasion Employed by Human Parasites. Front Cell Infect Microbiol 2021; 11:702125. [PMID: 34395313 PMCID: PMC8358743 DOI: 10.3389/fcimb.2021.702125] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/25/2021] [Indexed: 12/14/2022] Open
Abstract
For the establishment of a successful infection, i.e., long-term parasitism and a complete life cycle, parasites use various diverse mechanisms and factors, which they may be inherently bestowed with, or may acquire from the natural vector biting the host at the infection prelude, or may take over from the infecting host, to outmaneuver, evade, overcome, and/or suppress the host immunity, both innately and adaptively. This narrative review summarizes the up-to-date strategies exploited by a number of representative human parasites (protozoa and helminths) to counteract the target host immune defense. The revisited information should be useful for designing diagnostics and therapeutics as well as vaccines against the respective parasitic infections.
Collapse
Affiliation(s)
- Monrat Chulanetra
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wanpen Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
11
|
Evans CC, Burkman EJ, Dzimianski MT, Garner B, Moorhead AR. The Course of Brugia malayi Microfilaremia in Experimentally Infected Cats. Vector Borne Zoonotic Dis 2021; 21:586-592. [PMID: 34129397 DOI: 10.1089/vbz.2020.2761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
As one of the causative agents of lymphatic filariasis in humans, Brugia malayi has been established as the laboratory model of choice for studying this infection owing to its viability in small animal hosts, with the domestic cat being significant among these. The usefulness of individual feline infections is highly dependent on the levels of circulating microfilariae in the blood; thus, characterizing the course of microfilaremia benefits our understanding of this model. In B. malayi-endemic regions, cats are also known reservoirs of infection, and describing microfilaremia in a controlled setting may improve transmission modeling. We followed the course of B. malayi infection in 10 experimentally infected cats from inoculation to ultimate resolution. Seven cats developed patency, with a peak microfilaria concentration of 6525/mL. In addition, to identify cellular responses with potential value as predictors of patency, we measured the peripheral blood leukocyte counts during the first 8 months of infection and tested for correlations with lifelong microfilaria production. No strong relationships were observed, though cell values did appear to shift with the maturation phases of the parasite. The data we present reflect the course of microfilaremia in an important laboratory model under controlled conditions.
Collapse
Affiliation(s)
- Christopher C Evans
- Department of Infectious Diseases and College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Erica J Burkman
- Department of Infectious Diseases and College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Michael T Dzimianski
- Department of Infectious Diseases and College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Bridget Garner
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Andrew R Moorhead
- Department of Infectious Diseases and College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
12
|
Chen JY, Zhou JK, Pan W. Immunometabolism: Towards a Better Understanding the Mechanism of Parasitic Infection and Immunity. Front Immunol 2021; 12:661241. [PMID: 34122419 PMCID: PMC8191844 DOI: 10.3389/fimmu.2021.661241] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/13/2021] [Indexed: 12/26/2022] Open
Abstract
As a relatively successful pathogen, several parasites can establish long-term infection in host. This “harmonious symbiosis” status relies on the “precise” manipulation of host immunity and metabolism, however, the underlying mechanism is still largely elusive. Immunometabolism is an emerging crossed subject in recent years. It mainly discusses the regulatory mechanism of metabolic changes on reprogramming the key transcriptional and post-transcriptional events related to immune cell activation and effect, which provides a novel insight for understanding how parasites regulate the infection and immunity in hosts. The present study reviewed the current research progress on metabolic reprogramming mechanism exploited by parasites to modulate the function in various immune cells, highlighting the future exploitation of key metabolites or metabolic events to clarify the underlying mechanism of anti-parasite immunity and design novel intervention strategies against parasitic infection.
Collapse
Affiliation(s)
- Jing-Yue Chen
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,The First Clinical Medicine, Xuzhou Medical University, Xuzhou, China
| | - Ji-Kai Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,The First Clinical Medicine, Xuzhou Medical University, Xuzhou, China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
13
|
Ricciardi A, Bennuru S, Tariq S, Kaur S, Wu W, Elkahloun AG, Arakelyan A, Shaik J, Dorward DW, Nutman TB, Tolouei Semnani R. Extracellular vesicles released from the filarial parasite Brugia malayi downregulate the host mTOR pathway. PLoS Negl Trop Dis 2021; 15:e0008884. [PMID: 33411714 PMCID: PMC7790274 DOI: 10.1371/journal.pntd.0008884] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 10/13/2020] [Indexed: 12/21/2022] Open
Abstract
We have previously shown that the microfilarial (mf) stage of Brugia malayi can inhibit the mammalian target of rapamycin (mTOR; a conserved serine/threonine kinase critical for immune regulation and cellular growth) in human dendritic cells (DC) and we have proposed that this mTOR inhibition is associated with the DC dysfunction seen in filarial infections. Extracellular vesicles (EVs) contain many proteins and nucleic acids including microRNAs (miRNAs) that might affect a variety of intracellular pathways. Thus, EVs secreted from mf may elucidate the mechanism by which the parasite is able to modulate the host immune response during infection. EVs, purified from mf of Brugia malayi and confirmed by size through nanoparticle tracking analysis, were assessed by miRNA microarrays (accession number GSE157226) and shown to be enriched (>2-fold, p-value<0.05, FDR = 0.05) for miR100, miR71, miR34, and miR7. The microarray analysis compared mf-derived EVs and mf supernatant. After confirming their presence in EVs using qPCR for these miRNA targets, web-based target predictions (using MIRPathv3, TarBAse and MicroT-CD) predicted that miR100 targeted mTOR and its downstream regulatory protein 4E-BP1. Our previous data with live parasites demonstrated that mf downregulate the phosphorylation of mTOR and its downstream effectors. Additionally, our proteomic analysis of the mf-derived EVs revealed the presence of proteins commonly found in these vesicles (data are available via ProteomeXchange with identifier PXD021844). We confirmed internalization of mf-derived EVs by human DCs and monocytes using confocal microscopy and flow cytometry, and further demonstrated through flow cytometry, that mf-derived EVs downregulate the phosphorylation of mTOR in human monocytes (THP-1 cells) to the same degree that rapamycin (a known mTOR inhibitor) does. Our data collectively suggest that mf release EVs that interact with host cells, such as DC, to modulate host responses. Lymphatic filariasis, a neglected tropical disease caused by parasitic worms which affects millions of individuals, represents an important public health concern due to its high morbidity that significantly diminishes quality of life. The parasite is able to establish a chronic infection by manipulating its host’s immune responses. The larval mf stage of the parasite is of particular interest as the parasites are present in the peripheral blood and in constant contact with the host’s immune cells. We decided to investigate the role of mf-derived EVs in the modulation of human antigen presenting cells during infection. We showed that mf release EVs that are similar to exosomes, and these parasite vesicles are readily internalized by human DC. The mf-derived EVs possess a unique miRNA profile and are enriched in miRNAs that can target the mTOR pathway. We have also demonstrated that purified mf-derived EVs are capable of inhibiting mTOR signaling in human monocytes. Collectively, our results suggest that mf release exosome-like vesicles that modulate the immune metabolism of host antigen presenting cells.
Collapse
Affiliation(s)
- Alessandra Ricciardi
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, United States of America
- * E-mail:
| | - Sasisekhar Bennuru
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, United States of America
| | - Sameha Tariq
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, United States of America
| | - Sukhbir Kaur
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, United States of America
| | - Weiwei Wu
- Microarray Core, Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Abdel G. Elkahloun
- Microarray Core, Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland, United States of America
| | - Anush Arakelyan
- Section of Intracellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, United States of America
| | - Jahangheer Shaik
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, United States of America
| | - David W. Dorward
- RML Microscopy Unit, RML Research Technologies Section, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, Montana, United States of America
| | - Thomas B. Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, United States of America
| | - Roshanak Tolouei Semnani
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, United States of America
| |
Collapse
|
14
|
Joardar N, Mondal C, Sinha Babu SP. A review on the interactions between dendritic cells, filarial parasite and parasite-derived molecules in regulating the host immune responses. Scand J Immunol 2020; 93:e13001. [PMID: 33247468 DOI: 10.1111/sji.13001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/15/2020] [Accepted: 11/22/2020] [Indexed: 12/16/2022]
Abstract
Lymphatic filariasis (LF) is the second leading cause of parasitic disabilities that affects millions of people in India and several other tropical countries. The complexity of this disease is endorsed by various immunopathological consequences such as lymphangitis, lymphadenitis and elephantiasis. The immune evasion strategies that a filarial parasite usually follows are chiefly initiated with the communication between the invaded parasites and parasite-derived molecules, with the Toll-like receptors (TLRs) present on the surface of the antigen-presenting cells (APCs). Classically, the filarial parasites interact with the DCs resulting in lowering of CD4+ T-cell responses. These CD4+ T-cell responses are the key players behind the immune-mediated pathologies associated with LF. In chronic stage, the canonical pro-inflammatory immune responses are shifted towards an anti-inflammatory subtype, which is favouring the parasite survivability within the host. The central theme of this review article is to present the overall immune response elicited when an APC, particularly a DC, encounters a filarial parasite.
Collapse
Affiliation(s)
- Nikhilesh Joardar
- Parasitology Laboratory, Department of Zoology, Siksha-Bhavana, Visva-Bharati University, Santiniketan, India
| | - Chandrani Mondal
- Parasitology Laboratory, Department of Zoology, Siksha-Bhavana, Visva-Bharati University, Santiniketan, India
| | - Santi P Sinha Babu
- Parasitology Laboratory, Department of Zoology, Siksha-Bhavana, Visva-Bharati University, Santiniketan, India
| |
Collapse
|
15
|
Vanhamme L, Souopgui J, Ghogomu S, Ngale Njume F. The Functional Parasitic Worm Secretome: Mapping the Place of Onchocerca volvulus Excretory Secretory Products. Pathogens 2020; 9:pathogens9110975. [PMID: 33238479 PMCID: PMC7709020 DOI: 10.3390/pathogens9110975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 01/15/2023] Open
Abstract
Nematodes constitute a very successful phylum, especially in terms of parasitism. Inside their mammalian hosts, parasitic nematodes mainly dwell in the digestive tract (geohelminths) or in the vascular system (filariae). One of their main characteristics is their long sojourn inside the body where they are accessible to the immune system. Several strategies are used by parasites in order to counteract the immune attacks. One of them is the expression of molecules interfering with the function of the immune system. Excretory-secretory products (ESPs) pertain to this category. This is, however, not their only biological function, as they seem also involved in other mechanisms such as pathogenicity or parasitic cycle (molting, for example). We will mainly focus on filariae ESPs with an emphasis on data available regarding Onchocerca volvulus, but we will also refer to a few relevant/illustrative examples related to other worm categories when necessary (geohelminth nematodes, trematodes or cestodes). We first present Onchocerca volvulus, mainly focusing on the aspects of this organism that seem relevant when it comes to ESPs: life cycle, manifestations of the sickness, immunosuppression, diagnosis and treatment. We then elaborate on the function and use of ESPs in these aspects.
Collapse
Affiliation(s)
- Luc Vanhamme
- Department of Molecular Biology, Institute of Biology and Molecular Medicine, IBMM, Université Libre de Bruxelles, Rue des Professeurs Jeener et Brachet 12, 6041 Gosselies, Belgium; (J.S.); (F.N.N.)
- Correspondence:
| | - Jacob Souopgui
- Department of Molecular Biology, Institute of Biology and Molecular Medicine, IBMM, Université Libre de Bruxelles, Rue des Professeurs Jeener et Brachet 12, 6041 Gosselies, Belgium; (J.S.); (F.N.N.)
| | - Stephen Ghogomu
- Molecular and Cell Biology Laboratory, Biotechnology Unit, University of Buea, Buea P.O Box 63, Cameroon;
| | - Ferdinand Ngale Njume
- Department of Molecular Biology, Institute of Biology and Molecular Medicine, IBMM, Université Libre de Bruxelles, Rue des Professeurs Jeener et Brachet 12, 6041 Gosselies, Belgium; (J.S.); (F.N.N.)
- Molecular and Cell Biology Laboratory, Biotechnology Unit, University of Buea, Buea P.O Box 63, Cameroon;
| |
Collapse
|
16
|
Wang Y, Ehsan M, Wang S, Tian X, Yan R, Song X, Xu L, Li X. Modulatory functions of recombinant electron transfer flavoprotein α subunit protein from Haemonchus contortus on goat immune cells in vitro. Vet Parasitol 2020; 288:109300. [PMID: 33152677 DOI: 10.1016/j.vetpar.2020.109300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/25/2020] [Accepted: 10/26/2020] [Indexed: 01/13/2023]
Abstract
Suppression and modulation of the host immune response to parasitic nematodes have been extensively studied. In the present study, we cloned and produced recombinant electron transfer flavoprotein α subunit (ETFα) protein from Haemonchus contortus (rHCETFα), a parasitic nematode of small ruminants, and studied the effect of this protein on modulating the immune response of goat peripheral blood mononuclear cells (PBMCs) and monocytes. Immunohistochemical tests verified that the HCETFα protein was localized mainly in the intestinal wall and on the body surface of worms. Immunoblot analysis revealed that rHCETFα was recognized by the serum of goats artificially infected with H. contortus. Immunofluorescence analysis indicated that rHCETFα bound to the surface of PBMCs. rHCETFα was co-incubated with goat PBMCs to observe the immunomodulatory effects exerted by HCETFα on proliferation, apoptosis, cytokine secretion and nitric oxide (NO) production. The results showed that rHCETFα suppressed the proliferation of goat PBMCs stimulated by concanavalin A and induced apoptosis in goat PBMCs. After rHCETFα exposure, IL-2, IL-4, IL-17A and TNF-α expression was markedly reduced, whereas secretion of TGF-β1 was significantly elevated, in goat PBMCs. Moreover, rHCETFα up-regulated NO production in a dose-dependent manner. FITC-dextran internalization assays showed that rHCETFα inhibited phagocytosis of goat monocytes. These results elucidate the interaction between parasites and hosts at the molecular level, suggest a possible immunomodulatory target and contribute to the search for innovative proteins that may be candidate targets for drugs and vaccines.
Collapse
Affiliation(s)
- Yujian Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China; School of Life Science, Huizhou University, Huizhou 516007, PR China.
| | - Muhammad Ehsan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| | - Shuai Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| | - Xiaowei Tian
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| | - RuoFeng Yan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| | - XiaoKai Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| | - LiXin Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| | - XiangRui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
17
|
Wang Y, Ehsan M, Huang J, Aimulajiang K, Yan R, Song X, Xu L, Li X. Characterization of a rhodanese homologue from Haemonchus contortus and its immune-modulatory effects on goat immune cells in vitro. Parasit Vectors 2020; 13:454. [PMID: 32894178 PMCID: PMC7487571 DOI: 10.1186/s13071-020-04333-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 08/31/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Modulation of the host immune response by nematode parasites has been widely reported. Rhodaneses (thiosulfate: cyanide sulfurtransferases) are present in a wide range of organisms, such as archaea, bacteria, fungi, plants and animals. Previously, it was reported that a rhodanese homologue could be bound by goat peripheral blood mononuclear cells (PBMCs) in vivo. METHODS In the present study, we cloned and produced a recombinant rhodanese protein originating from Haemonchus contortus (rHCRD), a parasitic nematode of small ruminants. rHCRD was co-incubated with goat PBMCs to assess its immunomodulatory effects on proliferation, apoptosis and cytokine secretion. RESULTS We verified that the natural HCRD protein localized predominantly to the bowel wall and body surface of the parasite. We further demonstrated that serum produced by goats artificially infected with H. contortus successfully recognized rHCRD, which bound to goat PBMCs. rHCRD suppressed proliferation of goat PBMCs stimulated by concanavalin A but did not induce apoptosis in goat PBMCs. The production of TNF-α and IFN-γ decreased significantly, whereas secretion of IL-10 and TGF-β1 increased, in goat PBMCs after exposure to rHCRD. rHCRD also inhibited phagocytosis by goat monocytes. Moreover, rHCRD downregulated the expression of major histocompatibility complex (MHC)-II on goat monocytes in a dose-dependent manner, but did not alter MHC-I expression. CONCLUSIONS These results propose a possible immunomodulatory target that may help illuminate the interactions between parasites and their hosts at the molecular level and reveal innovative protein species as candidate drug and vaccine targets.
Collapse
Affiliation(s)
- Yujian Wang
- School of Life Science, Huizhou University, Huizhou, 516007, People's Republic of China.,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Muhammad Ehsan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Jianmei Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Kalibixiati Aimulajiang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - RuoFeng Yan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - XiaoKai Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - LiXin Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - XiangRui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| |
Collapse
|
18
|
Characterization of a phosphotyrosyl phosphatase activator homologue of the parasitic nematode Haemonchus contortus and its immunomodulatory effect on goat peripheral blood mononuclear cells in vitro. Int J Parasitol 2020; 50:1157-1166. [PMID: 32866490 DOI: 10.1016/j.ijpara.2020.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/25/2020] [Accepted: 07/02/2020] [Indexed: 01/06/2023]
Abstract
Suppression and modulation of the host immune response to parasitic nematodes have been extensively studied. In the present study, we cloned and produced recombinant phosphotyrosyl phosphatase activator protein from Haemonchus contortus (rHCPTPA), a parasitic nematode of small ruminants, and studied the effect of this protein on modulating the immune response of goat peripheral blood mononuclear cells. Enzymatic assays revealed that rHCPTPA enhanced the p-nitrophenylphosphate phosphatase activity of bovine PP2A1. Immunohistochemical tests verified that the HCPTPA protein was localised mainly in the bowel wall and on the body surface of worms. It was also shown that serum produced by goats artificially infected with H. contortus successfully recognised rHCPTPA, which conjugated with goat peripheral blood mononuclear cells. The rHCPTPA was then co-incubated with goat peripheral blood mononuclear cells to assess its immunomodulatory effects on proliferation, apoptosis, cytokine secretion, migration and nitric oxide production. Our results showed that rHCPTPA suppressed the proliferation of goat peripheral blood mononuclear cells stimulated by concanavalin A and induced apoptosis in goat peripheral blood mononuclear cells. After rHCPTPA exposure, IFN-γ and IL-2 expression was markedly reduced, whereas secretion of IL-10 and IL-4 was significantly elevated, in goat peripheral blood mononuclear cells. Moreover, rHCPTPA down-regulated nitric oxide production and migration of goat peripheral blood mononuclear cells in a dose-dependent manner. These results illuminate the interaction between parasites and hosts at the molecular level, suggest a possible immunomodulatory target and contribute to the search for innovative proteins that might be candidate targets for drugs and vaccines.
Collapse
|
19
|
Khatri V, Chauhan N, Kalyanasundaram R. Fecundity of adult female worms were affected when Brugia malayi infected Mongolian gerbils were immunized with a multivalent vaccine (rBmHAXT) against human lymphatic filarial parasite. Acta Trop 2020; 208:105487. [PMID: 32437645 PMCID: PMC7655632 DOI: 10.1016/j.actatropica.2020.105487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/18/2020] [Accepted: 04/01/2020] [Indexed: 01/03/2023]
Abstract
A multivalent recombinant fusion protein prophylactic vaccine, rBmHAXT developed against lymphatic filariasis (LF) demonstrated over 57% protection against challenge infection in rhesus macaque model. Currently, we do not know if the rBmHAXT vaccination has any effect on adult worms and/or on the fecundity of adult female worms. Thus, the major focus of this study was to determine the effect of rBmHAXT vaccination on Brugia malayi infected mongolian gerbils. We performed two sets of experiments. In the first set of experiment, gerbils were infected with 100 B. malayi L3. After confirming the establishment of infection, four rounds of DEC treatment and rBmHAXT vaccination was given. Results showed that following vaccination with rBmHAXT, the microfilaria (Mf) count was significantly decreased in all vaccinated animals compared to controls. At the end of these experiments, we collected and counted the established adult worms. There was a 36% reduction in the recovery of adult female worms, which might account for the low Mf load in vaccinated animals. In the second set of experiments, animals were vaccinated first with rBmHAXT followed by surgically implanting adult male or female B. malayi parasites into the peritoneal cavity to determine the effect of vaccination on each sex of the parasite. Our results show that the rBmHAXT vaccination has no effect on male adult worms compared to controls. However, there was 40% reduction in the Mf load in vaccinated animals that were transplanted with adult female worms. These findings suggested that the rBmHAXT vaccination has potential damaging effect on the fecundity of adult female worms. Scanning electron microscopy studies showed cuticular damage on the surface of adult female worms. These studies thus show that the rBmHAXT vaccination in infected gerbils has partial microfilaricidal effect and potentially affect the fecundity of adult female worms.
Collapse
Affiliation(s)
- Vishal Khatri
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, IL, USA
| | - Nikhil Chauhan
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, IL, USA
| | | |
Collapse
|
20
|
Mukherjee S, Karnam A, Das M, Babu SPS, Bayry J. Wuchereria bancrofti filaria activates human dendritic cells and polarizes T helper 1 and regulatory T cells via toll-like receptor 4. Commun Biol 2019; 2:169. [PMID: 31098402 PMCID: PMC6505026 DOI: 10.1038/s42003-019-0392-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 03/19/2019] [Indexed: 12/13/2022] Open
Abstract
Interaction between innate immune cells and parasite plays a key role in the immunopathogenesis of lymphatic filariasis. Despite being professional antigen presenting cells critical for the pathogen recognition, processing and presenting the antigens for mounting T cell responses, the dendritic cell response and its role in initiating CD4+ T cell response to filaria, in particular Wuchereria bancrofti, the most prevalent microfilaria is still not clear. Herein, we demonstrate that a 70 kDa phosphorylcholine-binding W. bancrofti sheath antigen induces human dendritic cell maturation and secretion of several pro-inflammatory cytokines. Further, microfilarial sheath antigen-stimulated dendritic cells drive predominantly Th1 and regulatory T cell responses while Th17 and Th2 responses are marginal. Mechanistically, sheath antigen-induced dendritic cell maturation, and Th1 and regulatory T cell responses are mediated via toll-like receptor 4 signaling. Our data suggest that W. bancrofti sheath antigen exploits dendritic cells to mediate distinct CD4+ T cell responses and immunopathogenesis of lymphatic filariasis.
Collapse
MESH Headings
- Animals
- Antigen Presentation
- Antigens, Helminth/genetics
- Antigens, Helminth/immunology
- Antigens, Helminth/pharmacology
- Cell Differentiation
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/parasitology
- Elephantiasis, Filarial/genetics
- Elephantiasis, Filarial/immunology
- Elephantiasis, Filarial/parasitology
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/immunology
- Gene Expression Regulation
- Host-Parasite Interactions/immunology
- Humans
- Immunity, Innate
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Interleukin-17/genetics
- Interleukin-17/immunology
- Lymphocyte Activation
- Microfilariae/genetics
- Microfilariae/immunology
- Microfilariae/pathogenicity
- Signal Transduction
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/parasitology
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Th1 Cells/parasitology
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Th17 Cells/parasitology
- Th2 Cells/drug effects
- Th2 Cells/immunology
- Th2 Cells/parasitology
- Toll-Like Receptor 4/genetics
- Toll-Like Receptor 4/immunology
- Wuchereria bancrofti/genetics
- Wuchereria bancrofti/immunology
- Wuchereria bancrofti/pathogenicity
Collapse
Affiliation(s)
- Suprabhat Mukherjee
- Department of Zoology (Centre for Advanced Studies), Visva-Bharati University, Santiniketan, 731235 India
- Institut National de la Santé et de la Recherche Médicale; Centre de Recherche des Cordeliers, Equipe—Immunopathologie et immuno-intervention thérapeutique, Sorbonne Universités, F-75006 Paris, France
- Present Address: Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal 713 340 India
| | - Anupama Karnam
- Institut National de la Santé et de la Recherche Médicale; Centre de Recherche des Cordeliers, Equipe—Immunopathologie et immuno-intervention thérapeutique, Sorbonne Universités, F-75006 Paris, France
| | - Mrinmoy Das
- Institut National de la Santé et de la Recherche Médicale; Centre de Recherche des Cordeliers, Equipe—Immunopathologie et immuno-intervention thérapeutique, Sorbonne Universités, F-75006 Paris, France
| | - Santi P. Sinha Babu
- Department of Zoology (Centre for Advanced Studies), Visva-Bharati University, Santiniketan, 731235 India
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale; Centre de Recherche des Cordeliers, Equipe—Immunopathologie et immuno-intervention thérapeutique, Sorbonne Universités, F-75006 Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, F-75006 Paris, France
| |
Collapse
|
21
|
Buerfent BC, Gölz L, Hofmann A, Rühl H, Stamminger W, Fricker N, Hess T, Oldenburg J, Nöthen MM, Schumacher J, Hübner MP, Hoerauf A. Transcriptome-wide analysis of filarial extract-primed human monocytes reveal changes in LPS-induced PTX3 expression levels. Sci Rep 2019; 9:2562. [PMID: 30796272 PMCID: PMC6385373 DOI: 10.1038/s41598-019-38985-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 01/15/2019] [Indexed: 12/24/2022] Open
Abstract
Filarial nematodes modulate immune responses in their host to enable their survival and mediate protective effects against autoimmunity and allergies. In this study, we examined the immunomodulatory capacity of extracts from the human pathogenic filaria Brugia malayi (BmA) on human monocyte responses in a transcriptome-wide manner to identify associated pathways and diseases. As previous transcriptome studies often observed quiescent responses of innate cells to filariae, the potential of BmA to alter LPS driven responses was investigated by analyzing >47.000 transcripts of monocytes from healthy male volunteers stimulated with BmA, Escherichia coli LPS or a sequential stimulation of both. In comparison to ~2200 differentially expressed genes in LPS-only stimulated monocytes, only a limited number of differentially expressed genes were identified upon BmA priming before LPS re-stimulation with only PTX3↓ reaching statistical significance after correcting for multiple testing. Nominal significant differences were reached for metallothioneins↑, MMP9↑, CXCL5/ENA-78↑, CXCL6/GCP-2↑, TNFRSF21↓, and CCL20/MIP3α↓ and were confirmed by qPCR or ELISA. Flow cytometric analysis of activation markers revealed a reduced LPS-induced expression of HLA-DR and CD86 on BmA-primed monocytes as well as a reduced apoptosis of BmA-stimulated monocytes. While our experimental design does not allow a stringent extrapolation of our results to the development of filarial pathology, several genes that were identified in BmA-primed monocytes had previously been associated with filarial pathology, supporting the need for further research.
Collapse
Affiliation(s)
- B C Buerfent
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Center for Human Genetics, University Hospital of Marburg, Marburg, Germany
| | - L Gölz
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, University of Bonn, Bonn, Germany
- Department of Orthodontics and Orofacial Orthopedics, University Hospital of Erlangen, Erlangen, Germany
| | - A Hofmann
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - H Rühl
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital of Bonn, Bonn, Germany
| | - W Stamminger
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
| | - N Fricker
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - T Hess
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Center for Human Genetics, University Hospital of Marburg, Marburg, Germany
| | - J Oldenburg
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital of Bonn, Bonn, Germany
| | - M M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
| | - J Schumacher
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Center for Human Genetics, University Hospital of Marburg, Marburg, Germany
| | - M P Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany.
| | - A Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital of Bonn, Bonn, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| |
Collapse
|
22
|
Bonne-Année S, Nutman TB. Human innate lymphoid cells (ILCs) in filarial infections. Parasite Immunol 2018; 40:10.1111/pim.12442. [PMID: 28504838 PMCID: PMC5685925 DOI: 10.1111/pim.12442] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/11/2017] [Indexed: 12/22/2022]
Abstract
Filarial infections are characteristically chronic and can cause debilitating diseases governed by parasite-induced innate and adaptive immune responses. Filarial parasites traverse or establish niches in the skin (migrating infective larvae), in nonmucosal tissues (adult parasite niche) and in the blood or skin (circulating microfilariae) where they intersect with the host immune response. While several studies have demonstrated that filarial parasites and their antigens can modulate myeloid cells (monocyte, macrophage and dendritic cell subsets), T- and B-lymphocytes and skin resident cell populations, the role of innate lymphoid cells during filarial infections has only recently emerged. Despite the identification and characterization of innate lymphoid cells (ILCs) in murine helminth infections, little is actually known about the role of human ILCs during parasitic infections. The focus of this review will be to highlight the composition of ILCs in the skin, lymphatics and blood; where the host-parasite interaction is well-defined and to examine the role of ILCs during filarial infections.
Collapse
Affiliation(s)
- S Bonne-Année
- Laboratory of Parasitic Diseases, Helminth Immunology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - T B Nutman
- Laboratory of Parasitic Diseases, Helminth Immunology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
23
|
Venugopal G, Mueller M, Hartmann S, Steinfelder S. Differential immunomodulation in human monocytes versus macrophages by filarial cystatin. PLoS One 2017; 12:e0188138. [PMID: 29141050 PMCID: PMC5687743 DOI: 10.1371/journal.pone.0188138] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 11/01/2017] [Indexed: 12/19/2022] Open
Abstract
Parasitic nematodes have evolved powerful immunomodulatory molecules to enable their survival in immunocompetent hosts by subverting immune responses and minimizing pathological processes. One filarial molecule known to counteract host immune responses by inducing IL-10 and regulatory macrophages in mice is filarial cystatin. During a patent filarial infection monocytes encounter microfilariae in the blood, an event that occurs in asymptomatically infected filariasis patients that are immunologically hyporeactive. The microfilarial larval stage was formerly shown to induce human regulatory monocytes and macrophages. Thus, here we aim was to determine how filarial cystatin of the human pathogenic filaria Brugia malayi (BmCPI-2) contributes to immune hyporesponsiveness in human monocytes and macrophages elicited by microfilaria. For this purpose, filarial cystatin was depleted from microfilarial lysate (Mf). Detecting the immunomodulatory potential of cystatin-depleted Mf revealed that IL-10, but not IL-8 and IL-6 induction in monocytes and macrophages is dependent on the presence of cystatin. In addition, the Mf-induced expression of the regulatory surface markers PD-L1 and PD-L2 in human monocytes, but not in macrophages, is dependent on cystatin. While Mf-treated monocytes result in decreased CD4+ T-cell proliferation in a co-culture assay, stimulation of T-cells with human monocytes treated with cystatin-depleted Mf lead to a restoration of CD4+ T-cell proliferation. Moreover, IL-10 induction by cystatin within Mf was dependent on p38 and ERK in macrophages, but independent of the ERK pathway in monocytes. These findings indicate that filarial nematodes differentially trigger and exploit various signaling pathways to induce immunomodulation in different myeloid cell subsets.
Collapse
Affiliation(s)
- Gopinath Venugopal
- Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Marion Mueller
- Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Susanne Hartmann
- Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
- * E-mail:
| | - Svenja Steinfelder
- Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
24
|
Grubor NM, Jovanova-Nesic KD, Shoenfeld Y. Liver cystic echinococcosis and human host immune and autoimmune follow-up: A review. World J Hepatol 2017; 9:1176-1189. [PMID: 29109850 PMCID: PMC5666304 DOI: 10.4254/wjh.v9.i30.1176] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 08/28/2017] [Accepted: 09/14/2017] [Indexed: 02/06/2023] Open
Abstract
Cystic echinococcosis (CE) is an infectious disease caused by the larvae of parasite Echinococcus granulosus (E. granulosus). To successfully establish an infection, parasite release some substances and molecules that can modulate host immune functions, stimulating a strong anti-inflammatory reaction to carry favor to host and to reserve self-survival in the host. The literature was reviewed using MEDLINE, and an open access search for immunology of hydatidosis was performed. Accumulating data from animal experiments and human studies provided us with exciting insights into the mechanisms involved that affect all parts of immunity. In this review we used the existing scientific data and discuss how these findings assisted with a better understanding of the immunology of E. granulosus infection in man. The aim of this study is to point the several facts that challenge immune and autoimmune responses to protect E. granulosus from elimination and to minimize host severe pathology. Understanding the immune mechanisms of E. granulosus infection in an intermediate human host will provide, we believe, a more useful treatment with immunomodulating molecules and possibly better protection from parasitic infections. Besides that, the diagnosis of CE has improved due to the application of a new molecular tool for parasite identification by using of new recombinant antigens and immunogenic peptides. More studies for the better understanding of the mechanisms of parasite immune evasion is necessary. It will enable a novel approach in protection, detection and improving of the host inflammatory responses. In contrast, according to the "hygiene hypothesis", clinical applications that decrease the incidence of infection in developed countries and recently in developing countries are at the origin of the increasing incidence of both allergic and autoimmune diseases. Thus, an understanding of the immune mechanisms of E. granulosus infection is extremely important.
Collapse
Affiliation(s)
- Nikica M Grubor
- Department of Hepatobiliary and Pancreatic Surgery, First Surgical University Hospital, Clinical Center of Serbia, School of Medicine University of Belgrade, 11000 Belgrade, Serbia
| | - Katica D Jovanova-Nesic
- Immunology Research Center, Institute of Virology, Vaccine and Sera-Torlak, 11221 Belgrade, Serbia
- European Center for Peace and Development, University for Peace in the United Nation established in Belgrade, 11000 Belgrade, Serbia.
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Aviv University, 5265601 Tel-Hashomer, Tel Aviv, Israel
| |
Collapse
|
25
|
Aravindhan V, Anand G. Cell Type-Specific Immunomodulation Induced by Helminthes: Effect on Metainflammation, Insulin Resistance and Type-2 Diabetes. Am J Trop Med Hyg 2017; 97:1650-1661. [PMID: 29141759 DOI: 10.4269/ajtmh.17-0236] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Recent epidemiological studies have documented an inverse relationship between the decreasing prevalence of helminth infections and the increasing prevalence of metabolic diseases ("metabolic hygiene hypothesis"). Chronic inflammation leading to insulin resistance (IR) has now been identified as a major etiological factor for a variety of metabolic diseases other than obesity and Type-2 diabetes (metainflammation). One way by which helminth infections such as filariasis can modulate IR is by inducing a chronic, nonspecific, low-grade, immune suppression mediated by modified T-helper 2 (Th2) response (induction of both Th2 and regulatory T cells) which can in turn suppress the proinflammatory responses and promote insulin sensitivity (IS). This article provides evidence on how the cross talk between the innate and adaptive arms of the immune responses can modulate IR/sensitivity. The cross talk between innate (macrophages, dendritic cells, natural killer cells, natural killer T cells, myeloid derived suppressor cells, innate lymphoid cells, basophils, eosinophils, and neutrophils) and adaptive (helper T [CD4+] cells, cytotoxic T [CD8+] cells and B cells) immune cells forms two opposing circuits, one associated with IR and the other associated with IS under the conditions of metabolic syndrome and helminth-mediated immunomodulation, respectively.
Collapse
|
26
|
Mishra R, Panda SK, Sahoo PK, Bal MS, Satapathy AK. Increased Fas ligand expression of peripheral B-1 cells correlated with CD4+T-cell apoptosis in filarial-infected patients. Parasite Immunol 2017; 39. [DOI: 10.1111/pim.12421] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 02/09/2017] [Indexed: 01/09/2023]
Affiliation(s)
- R. Mishra
- Regional Medical Research Centre (ICMR); Bhubaneswar Odisha India
| | - S. K. Panda
- Institutes of Life Sciences (DBT); Bhubaneswar Odisha India
| | - P. K. Sahoo
- Regional Medical Research Centre (ICMR); Bhubaneswar Odisha India
| | - M. S. Bal
- Regional Medical Research Centre (ICMR); Bhubaneswar Odisha India
| | - A. K. Satapathy
- Regional Medical Research Centre (ICMR); Bhubaneswar Odisha India
| |
Collapse
|
27
|
Kwarteng A, Ahuno ST. Immunity in Filarial Infections: Lessons from Animal Models and Human Studies. Scand J Immunol 2017; 85:251-257. [DOI: 10.1111/sji.12533] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 01/27/2017] [Indexed: 02/03/2023]
Affiliation(s)
- A. Kwarteng
- Department of Biochemistry and Biotechnology; Kwame Nkrumah University of Science Technology, PMB; Kumasi Ghana
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR); KNUST, PMB; Kumasi Ghana
| | - S. T. Ahuno
- Department of Biochemistry and Biotechnology; Kwame Nkrumah University of Science Technology, PMB; Kumasi Ghana
| |
Collapse
|
28
|
Vendelova E, Hrčková G, Lutz MB, Brehm K, Nono JK. In vitro culture of Mesocestoides corti metacestodes and isolation of immunomodulatory excretory-secretory products. Parasite Immunol 2017; 38:403-13. [PMID: 27120409 DOI: 10.1111/pim.12327] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 04/22/2016] [Indexed: 12/25/2022]
Abstract
Cestode-mediated diseases hold the interesting feature of persisting metacestode larvae dwelling within the host tissues, in the midst of the immune response. Excretory-secretory (ES) products of the metacestode larval stage modulate the host immune response and modify the outcome of the disease. Therefore, isolation and analysis of axenic metacestode ES products are crucial to study their properties. Here, we report the development of a system for long-term in vitro cultivation of the metacestode of the parasitic cestode Mesocestoides corti (syn. Mesocestoides vogae). Although feeder cells and host serum supported the early growth of the parasite, long-term survival was not dependent on host serum or host-derived factors enabling the collection of parasite released products in serum-free medium. Functionally, these axenic ES products recapitulated M. corti tetrathyridia's ability to inhibit LPS-driven IL-12p70 secretion by dendritic cells. Thus, our new axenic culture system will simplify the identification and characterization of M. corti-derived immunomodulatory factors that will indirectly enable the identification and characterization of corresponding factors in the metacestode larvae of medically relevant cestodes such as Echinococcus multilocularis that are not yet amenable to serum-free cultivation.
Collapse
Affiliation(s)
- E Vendelova
- Institute of Parasitology of the Slovak Academy of Sciences, Košice, Slovak Republic
| | - G Hrčková
- Institute of Parasitology of the Slovak Academy of Sciences, Košice, Slovak Republic
| | - M B Lutz
- Institute of Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - K Brehm
- Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - J K Nono
- Division of Immunology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Institute of Medical Research and Medicinal Plant Studies (IMPM), Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
| |
Collapse
|
29
|
Rodríguez E, Carasi P, Frigerio S, da Costa V, van Vliet S, Noya V, Brossard N, van Kooyk Y, García-Vallejo JJ, Freire T. Fasciola hepatica Immune Regulates CD11c + Cells by Interacting with the Macrophage Gal/GalNAc Lectin. Front Immunol 2017; 8:264. [PMID: 28360908 PMCID: PMC5350155 DOI: 10.3389/fimmu.2017.00264] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 02/23/2017] [Indexed: 01/03/2023] Open
Abstract
Fasciolosis, caused by Fasciola hepatica and Fasciola gigantica, is a trematode zoonosis of interest in public health and livestock production. Like other helminths, F. hepatica modulates the host immune response by inducing potent polarized Th2 and regulatory T cell immune responses and by downregulating the production of Th1 cytokines. In this work, we show that F. hepatica glycans increase Th2 immune responses by immunomodulating TLR-induced maturation and function of dendritic cells (DCs). This process was mediated by the macrophage Gal/GalNAc lectin (MGL) expressed on DCs, which recognizes the Tn antigen (GalNAc-Ser/Thr) on parasite components. More interestingly, we identified MGL-expressing CD11c+ cells in infected animals and showed that these cells are recruited both to the peritoneum and the liver upon F. hepatica infection. These cells express the regulatory cytokines IL-10, TNFα and TGFβ and a variety of regulatory markers. Furthermore, MGL+ CD11c+ cells expand parasite-specific Th2/regulatory cells and suppress Th1 polarization. The results presented here suggest a potential role of MGL in the immunomodulation of DCs induced by F. hepatica and contribute to a better understanding of the molecular and immunoregulatory mechanisms induced by this parasite.
Collapse
Affiliation(s)
- Ernesto Rodríguez
- Grupo de Inmunomodulación y Desarrollo de Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de La República , Montevideo , Uruguay
| | - Paula Carasi
- Grupo de Inmunomodulación y Desarrollo de Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de La República , Montevideo , Uruguay
| | - Sofía Frigerio
- Grupo de Inmunomodulación y Desarrollo de Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de La República , Montevideo , Uruguay
| | - Valeria da Costa
- Grupo de Inmunomodulación y Desarrollo de Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de La República , Montevideo , Uruguay
| | - Sandra van Vliet
- Department of Molecular Cell Biology and Immunology, VU University Medical Center , Amsterdam , Netherlands
| | - Verónica Noya
- Grupo de Inmunomodulación y Desarrollo de Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de La República , Montevideo , Uruguay
| | - Natalie Brossard
- Grupo de Inmunomodulación y Desarrollo de Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de La República , Montevideo , Uruguay
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, VU University Medical Center , Amsterdam , Netherlands
| | - Juan J García-Vallejo
- Department of Molecular Cell Biology and Immunology, VU University Medical Center , Amsterdam , Netherlands
| | - Teresa Freire
- Grupo de Inmunomodulación y Desarrollo de Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de La República , Montevideo , Uruguay
| |
Collapse
|
30
|
Anti-apoptotic effects of Sonic hedgehog signalling through oxidative stress reduction in astrocytes co-cultured with excretory-secretory products of larval Angiostrongylus cantonensis. Sci Rep 2017; 7:41574. [PMID: 28169282 PMCID: PMC5294578 DOI: 10.1038/srep41574] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 12/20/2016] [Indexed: 12/21/2022] Open
Abstract
Angiostrongylus cantonensis, the rat lungworm, is an important aetiologic agent of eosinophilic meningitis and meningoencephalitis in humans. Co-culturing astrocytes with soluble antigens of A. cantonensis activated the Sonic hedgehog (Shh) signalling pathway and inhibited the apoptosis of astrocytes via the activation of Bcl-2. This study was conducted to determine the roles of the Shh signalling pathway, apoptosis, and oxidative stress in astrocytes after treatment with excretory-secretory products (ESP) from A. cantonensis fifth-stage larvae. Although astrocyte viability was significantly decreased after ESP treatment, the expression of Shh signalling pathway related proteins (Shh, Ptch-1 and Gli-1) was significantly increased. However, apoptosis in astrocytes was significantly decreased after activation of the Shh signalling pathway. Moreover, superoxide and hydrogen superoxide levels in astrocytes were significantly reduced after the activation of Shh pathway signalling due to increasing levels of the antioxidants catalase and superoxide dismutase. These findings indicate that the anti-apoptotic effects of the Shh signalling pathway in the astrocytes of mice infected with A. cantonensis are due to reduced levels of oxidative stress caused by the activation of antioxidants.
Collapse
|
31
|
McCoy CJ, Reaves BJ, Giguère S, Coates R, Rada B, Wolstenholme AJ. Human Leukocytes Kill Brugia malayi Microfilariae Independently of DNA-Based Extracellular Trap Release. PLoS Negl Trop Dis 2017; 11:e0005279. [PMID: 28045905 PMCID: PMC5234842 DOI: 10.1371/journal.pntd.0005279] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 01/13/2017] [Accepted: 12/21/2016] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Wuchereria bancrofti, Brugia malayi and Brugia timori infect over 100 million people worldwide and are the causative agents of lymphatic filariasis. Some parasite carriers are amicrofilaremic whilst others facilitate mosquito-based disease transmission through blood-circulating microfilariae (Mf). Recent findings, obtained largely from animal model systems, suggest that polymorphonuclear leukocytes (PMNs) contribute to parasitic nematode-directed type 2 immune responses. When exposed to certain pathogens PMNs release extracellular traps (NETs) in the form of chromatin loaded with various antimicrobial molecules and proteases. PRINCIPAL FINDINGS In vitro, PMNs expel large amounts of NETs that capture but do not kill B. malayi Mf. NET morphology was confirmed by fluorescence imaging of worm-NET aggregates labelled with DAPI and antibodies to human neutrophil elastase, myeloperoxidase and citrullinated histone H4. A fluorescent, extracellular DNA release assay was used to quantify and observe Mf induced NETosis over time. Blinded video analyses of PMN-to-worm attachment and worm survival during Mf-leukocyte co-culture demonstrated that DNase treatment eliminates PMN attachment in the absence of serum, autologous serum bolsters both PMN attachment and PMN plus peripheral blood mononuclear cell (PBMC) mediated Mf killing, and serum heat inactivation inhibits both PMN attachment and Mf killing. Despite the effects of heat inactivation, the complement inhibitor compstatin did not impede Mf killing and had little effect on PMN attachment. Both human PMNs and monocytes, but not lymphocytes, are able to kill B. malayi Mf in vitro and NETosis does not significantly contribute to this killing. Leukocytes derived from presumably parasite-naïve U.S. resident donors vary in their ability to kill Mf in vitro, which may reflect the pathological heterogeneity associated with filarial parasitic infections. CONCLUSIONS/SIGNIFICANCE Human innate immune cells are able to recognize, attach to and kill B. malayi microfilariae in an in vitro system. This suggests that, in vivo, the parasites can evade this ability, or that only some human hosts support an infection with circulating Mf.
Collapse
Affiliation(s)
- Ciaran J. McCoy
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States of America
| | - Barbara J. Reaves
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States of America
| | - Steeve Giguère
- Department of Large Animal Medicine, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
| | - Ruby Coates
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States of America
- Department of Biology & Biochemistry, University of Bath, Bath, United Kingdom
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
| | - Adrian J. Wolstenholme
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States of America
| |
Collapse
|
32
|
Functional Impairment of Murine Dendritic Cell Subsets following Infection with Infective Larval Stage 3 of Brugia malayi. Infect Immun 2016; 85:IAI.00818-16. [PMID: 27799335 DOI: 10.1128/iai.00818-16] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 10/28/2016] [Indexed: 01/18/2023] Open
Abstract
Filarial parasites cause functional impairment of host dendritic cells (DCs). However, the effects of early infection on individual DC subsets are not known. In this study, we infected BALB/c mice with infective stage 3 larvae of the lymphatic filarial parasite Brugia malayi (Bm-L3) and studied the effect on fluorescence-activated cell sorter (FACS)-sorted DC subsets. While myeloid DCs (mDCs) accumulated by day 3 postinfection (p.i.), lymphoid DCs (LDCs) and CD8+ plasmacytoid DCs (pDCs) peaked at day 7 p.i. in the spleens and mesenteric lymph nodes (mLNs) of infected mice. Increased tumor necrosis factor alpha (TNF-α) but reduced interleukin 12 (IL-12) and Toll-like receptor 4 (TLR4), -6, and -9 and reciprocal secretion of IL-4 and IL-10 were also observed across all DC subsets. Interestingly, Bm-L3 increased the expression of CD80 and CD86 across all DC subsets but decreased that of major histocompatibility complex class II (MHC-II) on mDCs and pDCs, resulting in their impaired antigen uptake and presentation capacities, but maximally attenuated the T-cell proliferation capacity of only mDCs. Furthermore, Bm-L3 increased phosphorylated p38 (p-p38), but not p-ERK, in mDCs and LDCs but downregulated them in pDCs, along with differential modulation of protein tyrosine phosphatases SHP-1, TCPTP, PTEN, and PTP1B across all DC subsets. Taken together, we report hitherto undocumented effects of early Bm-L3 infection on purified host DC subsets that lead to their functional impairment and attenuated host T-cell response.
Collapse
|
33
|
Microfilariae of Brugia malayi Inhibit the mTOR Pathway and Induce Autophagy in Human Dendritic Cells. Infect Immun 2016; 84:2463-72. [PMID: 27297394 DOI: 10.1128/iai.00174-16] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 09/08/2016] [Indexed: 01/04/2023] Open
Abstract
Immune modulation is a hallmark of patent filarial infection, including suppression of antigen-presenting cell function and downmodulation of filarial antigen-specific T cell responses. The mammalian target of rapamycin (mTOR) signaling pathway has been implicated in immune regulation, not only by suppressing T cell responses but also by regulating autophagy (through mTOR sensing amino acid availability). Global proteomic analysis (liquid chromatography-tandem mass spectrometry) of microfilaria (mf)-exposed monocyte-derived dendritic cells (DC) indicated that multiple components of the mTOR signaling pathway, including mTOR, eIF4A, and eIF4E, are downregulated by mf, suggesting that mf target this pathway for immune modulation in DC. Utilizing Western blot analysis, we demonstrate that similar to rapamycin (a known mTOR inhibitor), mf downregulate the phosphorylation of mTOR and its regulatory proteins, p70S6K1 and 4E-BP1, a process essential for DC protein synthesis. As active mTOR signaling regulates autophagy, we examined whether mf exposure alters autophagy-associated processes. mf-induced autophagy was reflected in marked upregulation of phosphorylated Beclin 1, known to play an important role in both autophagosome formation and autolysosome fusion, in induction of LC3II, a marker of autophagosome formation, and in induced degradation of p62, a ubiquitin-binding protein that aggregates protein in autophagosomes and is degraded upon autophagy that was reduced significantly by mf exposure and by rapamycin. Together, these results suggest that Brugia malayi mf employ mechanisms of metabolic modulation in DC to influence the regulation of the host immune response by downregulating mTOR signaling, resulting in increased autophagy. Whether this is a result of the parasite-secreted rapamycin homolog is currently under study.
Collapse
|
34
|
Rodrigo MB, Schulz S, Krupp V, Ritter M, Wiszniewsky K, Arndts K, Tamadaho RSE, Endl E, Hoerauf A, Layland LE. Patency of Litomosoides sigmodontis infection depends on Toll-like receptor 4 whereas Toll-like receptor 2 signalling influences filarial-specific CD4(+) T-cell responses. Immunology 2016; 147:429-42. [PMID: 26714796 DOI: 10.1111/imm.12573] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 12/18/2015] [Accepted: 12/21/2015] [Indexed: 12/14/2022] Open
Abstract
BALB/c mice develop a patent state [release of microfilariae (Mf), the transmission life-stage, into the periphery] when exposed to the rodent filariae Litomosoides sigmodontis. Interestingly, only a portion of the infected mice become patent, which reflects the situation in human individuals infected with Wuchereria bancrofti. Since those individuals had differing filarial-specific profiles, this study compared differences in immune responses between Mf(+) and Mf(-) infected BALB/c mice. We demonstrate that cultures of total spleen or mediastinal lymph node cells from Mf(+) mice produce significantly more interleukin-5 (IL-5) to filarial antigens but equal levels of IL-10 when compared with Mf(-) mice. However, isolated CD4(+) T cells from Mf(+) mice produced significantly higher amounts of all measured cytokines, including IL-10, when compared with CD4(+) T-cell responses from Mf(-) mice. Since adaptive immune responses are influenced by triggering the innate immune system we further studied the immune profiles and parasitology in infected Toll-like receptor-2-deficient (TLR2(-/-)) and TLR4(-/-) BALB/c mice. Ninety-three per cent of L. sigmodontis-exposed TLR4(-/-) BALB/c mice became patent (Mf(+)) although worm numbers remained comparable to those in Mf(+) wild-type controls. Lack of TLR2 had no influence on patency outcome or worm burden but infected Mf(+) mice had significantly lower numbers of Foxp3(+) regulatory T cells and dampened peripheral immune responses. Interestingly, in vitro culturing of CD4(+) T cells from infected wild-type mice with granulocyte-macrophage colony-stimulating factor-derived TLR2(-/-) dendritic cells resulted in an overall diminished cytokine profile to filarial antigens. Hence, triggering TLR4 or TLR2 during chronic filarial infection has a significant impact on patency and efficient CD4(+) T-cell responses, respectively.
Collapse
Affiliation(s)
- Maria B Rodrigo
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital of Bonn, Bonn, Germany
| | - Sandy Schulz
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital of Bonn, Bonn, Germany
| | - Vanessa Krupp
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital of Bonn, Bonn, Germany
| | - Manuel Ritter
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital of Bonn, Bonn, Germany
| | - Katharina Wiszniewsky
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital of Bonn, Bonn, Germany
| | - Kathrin Arndts
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital of Bonn, Bonn, Germany
| | - Ruth S E Tamadaho
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital of Bonn, Bonn, Germany
| | - Elmar Endl
- Institute for Molecular Medicine, University Hospital of Bonn, Bonn, Germany
| | - Achim Hoerauf
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital of Bonn, Bonn, Germany.,German Centre for Infection Research (DZIF), Partner Site, Bonn-Cologne, Bonn, Germany
| | - Laura E Layland
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital of Bonn, Bonn, Germany.,German Centre for Infection Research (DZIF), Partner Site, Bonn-Cologne, Bonn, Germany
| |
Collapse
|
35
|
Brugia malayi Antigen (BmA) Inhibits HIV-1 Trans-Infection but Neither BmA nor ES-62 Alter HIV-1 Infectivity of DC Induced CD4+ Th-Cells. PLoS One 2016; 11:e0146527. [PMID: 26808476 PMCID: PMC4726616 DOI: 10.1371/journal.pone.0146527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 12/19/2015] [Indexed: 11/19/2022] Open
Abstract
One of the hallmarks of HIV-1 disease is the association of heightened CD4+ T-cell activation with HIV-1 replication. Parasitic helminths including filarial nematodes have evolved numerous and complex mechanisms to skew, dampen and evade human immune responses suggesting that HIV-1 infection may be modulated in co-infected individuals. Here we studied the effects of two filarial nematode products, adult worm antigen from Brugia malayi (BmA) and excretory-secretory product 62 (ES-62) from Acanthocheilonema viteae on HIV-1 infection in vitro. Neither BmA nor ES-62 influenced HIV-1 replication in CD4+ enriched T-cells, with either a CCR5- or CXCR4-using virus. BmA, but not ES-62, had the capacity to bind the C-type lectin dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) thereby inhibiting HIV-1 trans-infection of CD4+ enriched T-cells. As for their effect on DCs, neither BmA nor ES-62 could enhance or inhibit DC maturation as determined by CD83, CD86 and HLA-DR expression, or the production of IL-6, IL-10, IL-12 and TNF-α. As expected, due to the unaltered DC phenotype, no differences were found in CD4+ T helper (Th) cell phenotypes induced by DCs treated with either BmA or ES-62. Moreover, the HIV-1 susceptibility of the Th-cell populations induced by BmA or ES-62 exposed DCs was unaffected for both CCR5- and CXCR4-using HIV-1 viruses. In conclusion, although BmA has the potential capacity to interfere with HIV-1 transmission or initial viral dissemination through preventing the virus from interacting with DCs, no differences in the Th-cell polarizing capacity of DCs exposed to BmA or ES-62 were observed. Neither antigenic source demonstrated beneficial or detrimental effects on the HIV-1 susceptibility of CD4+ Th-cells induced by exposed DCs.
Collapse
|
36
|
Nutman TB. Looking beyond the induction of Th2 responses to explain immunomodulation by helminths. Parasite Immunol 2015; 37:304-13. [PMID: 25869527 DOI: 10.1111/pim.12194] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 03/25/2015] [Indexed: 01/01/2023]
Abstract
Although helminth infections are characteristically associated with Th2-mediated responses that include the production of the prototypical cytokines IL-4, IL-5 and IL-13 by CD4(+) cells, the production of IgE, peripheral blood eosinophilia and mucus production in localized sites, these responses are largely attenuated when helminth infections become less acute. This modulation of the immune response that occurs with chronic helminth infection is often induced by molecules secreted by helminth parasites, by non-Th2 regulatory CD4(+) cells, and by nonclassical B cells, macrophages and dendritic cells. This review will focus on those parasite- and host-mediated mechanisms underlying the modulated T-cell response that occurs as the default in chronic helminth infections.
Collapse
Affiliation(s)
- T B Nutman
- Helminth Immunology Section, Laboratory of Parasitic Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
37
|
Sharma P, Sharma A, Vishwakarma AL, Agnihotri PK, Sharma S, Srivastava M. Host lung immunity is severely compromised during tropical pulmonary eosinophilia: role of lung eosinophils and macrophages. J Leukoc Biol 2015; 99:619-28. [PMID: 26489428 DOI: 10.1189/jlb.4a0715-309rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 10/01/2015] [Indexed: 12/28/2022] Open
Abstract
Eosinophils play a central role in the pathogenesis of tropical pulmonary eosinophilia, a rare, but fatal, manifestation of filariasis. However, no exhaustive study has been done to identify the genes and proteins of eosinophils involved in the pathogenesis of tropical pulmonary eosinophilia. In the present study, we established a mouse model of tropical pulmonary eosinophilia that mimicked filarial manifestations of human tropical pulmonary eosinophilia pathogenesis and used flow cytometry-assisted cell sorting and real-time RT-PCR to study the gene expression profile of flow-sorted, lung eosinophils and lung macrophages during tropical pulmonary eosinophilia pathogenesis. Our results show that tropical pulmonary eosinophilia mice exhibited increased levels of IL-4, IL-5, CCL5, and CCL11 in the bronchoalveolar lavage fluid and lung parenchyma along with elevated titers of IgE and IgG subtypes in the serum. Alveolar macrophages from tropical pulmonary eosinophilia mice displayed decreased phagocytosis, attenuated nitric oxide production, and reduced T-cell proliferation capacity, and FACS-sorted lung eosinophils from tropical pulmonary eosinophilia mice upregulated transcript levels of ficolin A and anti-apoptotic gene Bcl2,but proapoptotic genes Bim and Bax were downregulated. Similarly, flow-sorted lung macrophages upregulated transcript levels of TLR-2, TLR-6, arginase-1, Ym-1, and FIZZ-1 but downregulated nitric oxide synthase-2 levels, signifying their alternative activation. Taken together, we show that the pathogenesis of tropical pulmonary eosinophilia is marked by functional impairment of alveolar macrophages, alternative activation of lung macrophages, and upregulation of anti-apoptotic genes by eosinophils. These events combine together to cause severe lung inflammation and compromised lung immunity. Therapeutic interventions that can boost host immune response in the lungs might thus provide relief to patients with tropical pulmonary eosinophilia.
Collapse
Affiliation(s)
- Pankaj Sharma
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| | - Aditi Sharma
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| | - Achchhe Lal Vishwakarma
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| | - Promod Kumar Agnihotri
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| | - Sharad Sharma
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| | - Mrigank Srivastava
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| |
Collapse
|
38
|
Buerfent BC, Gondorf F, Wohlleber D, Schumak B, Hoerauf A, Hübner MP. Escherichia coli-induced immune paralysis is not exacerbated during chronic filarial infection. Immunology 2015; 145:150-60. [PMID: 25521437 DOI: 10.1111/imm.12435] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 11/26/2014] [Accepted: 12/12/2014] [Indexed: 01/17/2023] Open
Abstract
Sepsis initially starts with a systemic inflammatory response (SIRS phase) and is followed by a compensatory anti-inflammatory response syndrome (CARS) that causes impaired adaptive T-cell immunity, immune paralysis and an increased susceptibility to secondary infections. In contrast, parasitic filariae release thousands of microfilariae into the peripheral blood without triggering inflammation, as they induce regulatory, anti-inflammatory host responses. Hence, we investigated the impact of chronic filarial infection on adaptive T-cell responses during the SIRS and CARS phases of a systemic bacterial infection and analysed the development of T-cell paralysis following a subsequent adenovirus challenge in BALB/c mice. Chronic filarial infection impaired adenovirus-specific CD8(+) T-cell cytotoxicity and interferon-γ responses in the absence of a bacterial challenge and led to higher numbers of splenic CTLA-4(+) CD4(+) T cells, whereas splenic T-cell expression of CD69 and CD62 ligand, serum cytokine levels and regulatory T-cell frequencies were comparable to naive controls. Irrespective of filarial infection, the SIRS phase dominated 6-24 hr after intravenous Escherichia coli challenge with increased T-cell activation and pro-inflammatory cytokine production, whereas the CARS phase occurred 6 days post E. coli challenge and correlated with high levels of transforming growth factor-β and increased CD62 ligand T-cell expression. Escherichia coli-induced impairment of adenovirus-specific CD8(+) T-cell cytotoxicity and interferon-γ production was not additionally impaired by chronic filarial infection. This suggests that filarial immunoregulation does not exacerbate E. coli-induced T-cell paralysis.
Collapse
Affiliation(s)
- Benedikt C Buerfent
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital of Bonn, Bonn, Germany
| | | | | | | | | | | |
Collapse
|
39
|
Kamalakannan V, Shiny A, Babu S, Narayanan RB. Autophagy protects monocytes from Wolbachia heat shock protein 60-induced apoptosis and senescence. PLoS Negl Trop Dis 2015; 9:e0003675. [PMID: 25849993 PMCID: PMC4388636 DOI: 10.1371/journal.pntd.0003675] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Accepted: 03/05/2015] [Indexed: 12/17/2022] Open
Abstract
Monocyte dysfunction by filarial antigens has been a major mechanism underlying immune evasion following hyporesponsiveness during patent lymphatic filariasis. Recent studies have initiated a paradigm shift to comprehend the immunological interactions of Wolbachia and its antigens in inflammation, apoptosis, lymphocyte anergy, etc. Here we showed that recombinant Wolbachia heat shock protein 60 (rWmhsp60) interacts with TLR-4 and induces apoptosis in monocytes of endemic normal but not in chronic patients. Higher levels of reactive oxygen species (ROS) induced after TLR-4 stimulation resulted in loss of mitochondrial membrane potential and caspase cascade activation, which are the plausible reason for apoptosis. Furthermore, release in ROS owing to TLR-4 signaling resulted in the activation of NF-κB p65 nuclear translocation which leads to inflammation and apoptosis via TNF receptor pathway following the increase in IL-6 and TNF-α level. Here for the first time, we report that in addition to apoptosis, rWmhsp60 antigen in filarial pathogenesis also induces molecular senescence in monocytes. Targeting TLR-4, therefore, presents a promising candidate for treating rWmhsp60-induced apoptosis and senescence. Strikingly, induction of autophagy by rapamycin detains TLR-4 in late endosomes and subverts TLR-4-rWmhsp60 interaction, thus protecting TLR-4–mediated apoptosis and senescence. Furthermore, rapamycin-induced monocytes were unresponsive to rWmhsp60, and activated lymphocytes following PHA stimulation. This study demonstrates that autophagy mediates the degradation of TLR-4 signaling and protects monocytes from rWmhsp60 induced apoptosis and senescence. Despite knowing the significance of Wolbachia in helminth infections, our understanding of immunity and pathogenesis remains incomplete. Therefore, considering the gravity of the problem, the present study provides evidence that Wolbachia heat shock protein 60 induces apoptosis and senescence through TLR-4. Also, binding of rWmhsp60 to TLR-4 triggered caspase cascade activation following, ROS-mediated mitochondrial potential loss. Moreover, we found that nuclear translocation of NF-κB p65 was predominantly related to TLR-4 expression and resulted in apoptosis- and senescence-mediated inflammation via TNF-α and IL-6. Hence, we hypothesized that modifying TLR-4 expression may provide a plausible target for designing antiparasitic drugs. Here we have shown that induction of autophagy by rapamycin destabilizes TLR-4 expression and protects monocytes from rWmhsp60-induced apoptosis and senescence. In addition, rapamycin-induced monocytes were unresponsive to rWmhsp60 and triggered lymphocyte activation after PHA stimulation. Thus, synergistic usage of rapamycin with existing anti-filarial drugs might reduce the TLR-mediated inflammatory reactions following microfilaricidal treatment.
Collapse
Affiliation(s)
| | - Abijit Shiny
- Madras Diabetes Research Foundation and Dr. Mohan's Diabetes Specialties Centre, Chennai, Tamil Nadu, India
| | - Subash Babu
- National Institutes of Health—National Institute for Research in Tuberculosis-International Center for Excellence in Research, National Institute for Research in Tuberculosis, Chetpet, Chennai, Tamil Nadu, India
| | | |
Collapse
|
40
|
Chatterjee S, Nutman TB. Helminth-induced immune regulation: implications for immune responses to tuberculosis. PLoS Pathog 2015; 11:e1004582. [PMID: 25632943 PMCID: PMC4310592 DOI: 10.1371/journal.ppat.1004582] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Soumya Chatterjee
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Thomas B Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
41
|
Babu S, Nutman TB. Immunology of lymphatic filariasis. Parasite Immunol 2014; 36:338-46. [PMID: 24134686 DOI: 10.1111/pim.12081] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 10/11/2013] [Indexed: 12/13/2022]
Abstract
The immune responses to filarial parasites encompass a complex network of innate and adaptive cells whose interaction with the parasite underlies a spectrum of clinical manifestations. The predominant immunological feature of lymphatic filariasis is an antigen-specific Th2 response and an expansion of IL-10 producing CD4(+) T cells that is accompanied by a muted Th1 response. This antigen-specific T-cell hyporesponsiveness appears to be crucial for the maintenance of the sustained, long-standing infection often with high parasite densities. While the correlates of protective immunity to lymphatic filariasis are still incompletely understood, primarily due to the lack of suitable animal models to study susceptibility, it is clear that T cells and to a certain extent B cells are required for protective immunity. Host immune responses, especially CD4(+) T-cell responses clearly play a role in mediating pathological manifestations of LF, including lymphedema, hydrocele and elephantiasis. The main underlying defect in the development of clinical pathology appears to be a failure to induce T-cell hyporesponsiveness in the face of antigenic stimulation. Finally, another intriguing feature of filarial infections is their propensity to induce bystander effects on a variety of immune responses, including responses to vaccinations, allergens and to other infectious agents. The complexity of the immune response to filarial infection therefore provides an important gateway to understanding the regulation of immune responses to chronic infections, in general.
Collapse
Affiliation(s)
- S Babu
- NIAID-NIRT-ICER, Chennai, India
| | | |
Collapse
|
42
|
O'Regan NL, Steinfelder S, Venugopal G, Rao GB, Lucius R, Srikantam A, Hartmann S. Brugia malayi microfilariae induce a regulatory monocyte/macrophage phenotype that suppresses innate and adaptive immune responses. PLoS Negl Trop Dis 2014; 8:e3206. [PMID: 25275395 PMCID: PMC4183501 DOI: 10.1371/journal.pntd.0003206] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 08/21/2014] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Monocytes and macrophages contribute to the dysfunction of immune responses in human filariasis. During patent infection monocytes encounter microfilariae in the blood, an event that occurs in asymptomatically infected filariasis patients that are immunologically hyporeactive. AIM To determine whether blood microfilariae directly act on blood monocytes and in vitro generated macrophages to induce a regulatory phenotype that interferes with innate and adaptive responses. METHODOLOGY AND PRINCIPAL FINDINGS Monocytes and in vitro generated macrophages from filaria non-endemic normal donors were stimulated in vitro with Brugia malayi microfilarial (Mf) lysate. We could show that monocytes stimulated with Mf lysate develop a defined regulatory phenotype, characterised by expression of the immunoregulatory markers IL-10 and PD-L1. Significantly, this regulatory phenotype was recapitulated in monocytes from Wuchereria bancrofti asymptomatically infected patients but not patients with pathology or endemic normals. Monocytes from non-endemic donors stimulated with Mf lysate directly inhibited CD4+ T cell proliferation and cytokine production (IFN-γ, IL-13 and IL-10). IFN-γ responses were restored by neutralising IL-10 or PD-1. Furthermore, macrophages stimulated with Mf lysate expressed high levels of IL-10 and had suppressed phagocytic abilities. Finally Mf lysate applied during the differentiation of macrophages in vitro interfered with macrophage abilities to respond to subsequent LPS stimulation in a selective manner. CONCLUSIONS AND SIGNIFICANCE Conclusively, our study demonstrates that Mf lysate stimulation of monocytes from healthy donors in vitro induces a regulatory phenotype, characterized by expression of PD-L1 and IL-10. This phenotype is directly reflected in monocytes from filarial patients with asymptomatic infection but not patients with pathology or endemic normals. We suggest that suppression of T cell functions typically seen in lymphatic filariasis is caused by microfilaria-modulated monocytes in an IL-10-dependent manner. Together with suppression of macrophage innate responses, this may contribute to the overall down-regulation of immune responses observed in asymptomatically infected patients.
Collapse
Affiliation(s)
- Noëlle Louise O'Regan
- Freie Universität Berlin, Center for Infection Medicine, Institute of Immunology, Berlin, Germany
| | - Svenja Steinfelder
- Freie Universität Berlin, Center for Infection Medicine, Institute of Immunology, Berlin, Germany
| | - Gopinath Venugopal
- Freie Universität Berlin, Center for Infection Medicine, Institute of Immunology, Berlin, Germany
| | - Gopala B. Rao
- Blue Peter Public Health and Research Centre-LEPRA Society, Hyderabad, Andhra Pradesh, India
| | - Richard Lucius
- Humboldt Universität Berlin, Department of Biology, Molecular Parasitology, Berlin, Germany
| | - Aparna Srikantam
- Blue Peter Public Health and Research Centre-LEPRA Society, Hyderabad, Andhra Pradesh, India
| | - Susanne Hartmann
- Freie Universität Berlin, Center for Infection Medicine, Institute of Immunology, Berlin, Germany
- * E-mail:
| |
Collapse
|
43
|
Das Mohapatra A, Panda SK, Pradhan AK, Prusty BK, Satapathy AK, Ravindran B. Filarial antigens mediate apoptosis of human monocytes through Toll-like receptor 4. J Infect Dis 2014; 210:1133-44. [PMID: 24737802 DOI: 10.1093/infdis/jiu208] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Apoptosis of several host cells induced by parasites/parasite products has been investigated in human filariasis to understand immune hyporesponsiveness. However, apoptosis of monocytes-one of the major antigen presenting cells in peripheral circulation, which are chronically exposed to filarial antigens in infected subjects-is yet to be understood. METHODS Apoptosis of human monocytes with Brugia pahangi antigen (BpA) was demonstrated by scoring several apoptotic markers using flow cytometry. Ability of BpA and plasma of infected subjects to suppress lymphocyte proliferation was demonstrated by (3)H thymidine incorporation assay and carboxyfluorescein succinimidyl ester dilution assay. RESULTS BpA induced significant apoptosis of normal human monocytes, primarily through Toll-like receptor 4 (TLR4), and suppressed phytohemagglutinin (PHA)-mediated proliferation of normal human T lymphocytes. However, monocytes of Wuchereria bancrofti-infected subjects were resistant to BpA-induced apoptosis. Plasma of infected subjects also mediated apoptosis of normal monocytes, presumably due to circulating filarial antigens, and resulted in inhibition of PHA-induced proliferation. CONCLUSION Normal human monocytes were found to be qualitatively different from those of filariasis-infected subjects; whereas filarial antigens mediate apoptosis of normal human monocytes through TLR4, those of infected subjects were found to be resistant.
Collapse
Affiliation(s)
| | | | | | | | - Ashok Kumar Satapathy
- Department of Immunology, Regional Medical Research Center, Indian Council of Medical Research, Bhubaneswar, India
| | | |
Collapse
|
44
|
Hübner MP, Layland LE, Hoerauf A. Helminths and their implication in sepsis - a new branch of their immunomodulatory behaviour? Pathog Dis 2013; 69:127-41. [PMID: 23929557 PMCID: PMC4285315 DOI: 10.1111/2049-632x.12080] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 07/24/2013] [Accepted: 07/31/2013] [Indexed: 01/04/2023] Open
Abstract
The prevalence of autoimmune and allergic disorders has dramatically increased in developed countries, and it is believed that our ‘cleaner living’ reduces exposure to certain microorganisms and leads to deviated and/or reduced regulation of the immune system. In substantiation of this health hygiene hypothesis, multiple epidemiological studies and animal models have characterized the protective immune responses induced by helminths during auto-inflammatory disorders. The beneficial effects of such helminths, like schistosomes and filariae, are thought to lie in their immunomodulatory capacity, which can be induced by different life-cycle stages or components thereof. In addition to suppressing autoimmunity recent evidence indicates that concurrent helminth infections also counterbalance exacerbated pro-inflammatory immune responses that occur during sepsis, improving survival. As with allergy, epidemiological studies have observed a steady rise in severe sepsis cases and although this may have resulted from several factors (immunosuppressive drugs, chemotherapy, transplantation, increased awareness and increased surgical procedures), it is tempting to hypothesize that the lack of helminth infections in Western countries may have contributed to this phenomenon. This review summarizes how helminths modulate host immunity during sepsis, such as manipulating macrophage activation and provides an overview about the possible implications that may arise during overwhelming bacterial co-infections. This well written review gives a comprehensive overview on the immunopathology of sepsis and the modulation of immune responses by helminths. It provides evidence that helminths or components thereof may improve the outcome of severe infections. This will allow the development of therapeutic strategies to fight infections and sepsis.
Collapse
Affiliation(s)
- Marc P Hübner
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn, Bonn, Germany
| | | | | |
Collapse
|
45
|
White RR, Artavanis-Tsakonas K. How helminths use excretory secretory fractions to modulate dendritic cells. Virulence 2012; 3:668-77. [PMID: 23221477 PMCID: PMC3545949 DOI: 10.4161/viru.22832] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
It is well known that helminth parasites have immunomodulatory effects on their hosts. They characteristically cause a skew toward TH2 immunity, stimulate Treg cells while simultaneously inhibiting TH1 and TH17 responses. Additionally, they induce eosinophilia and extensive IgE release. The exact mechanism of how the worms achieve this effect have yet to be fully elucidated; however, parasite-derived secretions and their interaction with antigen presenting cells have been centrally implicated. Herein, we will review the effects of helminth excretory-secretory fractions on dendritic cells and discuss how this interaction is crucial in shaping the host response.
Collapse
Affiliation(s)
- Rhiannon R White
- Department of Life Sciences, Division of Cell and Molecular Biology, Imperial College London, London, UK
| | | |
Collapse
|
46
|
Immunopathogenesis of lymphatic filarial disease. Semin Immunopathol 2012; 34:847-61. [PMID: 23053393 DOI: 10.1007/s00281-012-0346-4] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 09/13/2012] [Indexed: 01/06/2023]
Abstract
Although two thirds of the 120 million people infected with lymph-dwelling filarial parasites have subclinical infections, ~40 million have lymphedema and/or other pathologic manifestations including hydroceles (and other forms of urogenital disease), episodic adenolymphangitis, tropical pulmonary eosinophilia, lymphedema, and (in its most severe form) elephantiasis. Adult filarial worms reside in the lymphatics and lymph nodes and induce changes that result in dilatation of lymphatics and thickening of the lymphatic vessel walls. Progressive lymphatic damage and pathology results from the summation of the effect of tissue alterations induced by both living and nonliving adult parasites, the host inflammatory response to the parasites and their secreted antigens, the host inflammatory response to the endosymbiont Wolbachia, and those seen as a consequence of secondary bacterial or fungal infections. Inflammatory damage induced by filarial parasites appears to be multifactorial, with endogenous parasite products, Wolbachia, and host immunity all playing important roles. This review will initially examine the prototypical immune responses engendered by the parasite and delineate the regulatory mechanisms elicited to prevent immune-mediated pathology. This will be followed by a discussion of the proposed mechanisms underlying pathogenesis, with the central theme being that pathogenesis is a two-step process-the first initiated by the parasite and host innate immune system and the second propagated mainly by the host's adaptive immune system and by other factors (including secondary infections).
Collapse
|
47
|
Sreenivas K, Vijayan K, Babu S, Narayanan RB. Recombinant Brugia malayi pepsin inhibitor (rBm33) induced monocyte function and absence of apoptotic cell death: an in vitro study. Microb Pathog 2012; 53:19-27. [PMID: 22484090 PMCID: PMC3512105 DOI: 10.1016/j.micpath.2012.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Revised: 03/19/2012] [Accepted: 03/26/2012] [Indexed: 10/28/2022]
Abstract
The effect of recombinant Brugia malayi pepsin inhibitor (rBm33) on human monocytes/macrophages has been examined using THP-1 cells. THP-1 cells stimulated with rBm33 showed enhanced levels of expression of pro-inflammatory cytokines (IL-1β, TNF-α, IL-6) and diminished levels of IL-12, iNOS and anti-inflammatory cytokine (IL-10) expression suggesting the predominant features of Th1 response. Phorbol-12-myristate-13-acetate (PMA) treated THP-1 cells stimulated with rBm33 and subsequent incubation with GFP expressing Escherichia coli (E. coli) for 2 h enhanced the uptake of E. coli. Nitric oxide (NO) levels measured in the supernatants of these cultures did not show significant changes. Apoptotic studies with Peripheral Blood Mononuclear Cells (PBMCs) from normal individuals stimulated with rBm33 did not induce apoptosis of monocytes or lymphocytes. These observations suggest that rBm33 stimulates macrophages to induce Th1 response and does not promote apoptosis.
Collapse
Affiliation(s)
- Kirthika Sreenivas
- Centre for Biotechnology, Anna University, S.P road, Guindy, Chennai, Tamil Nadu 600025, India
| | - Kamalakannan Vijayan
- Centre for Biotechnology, Anna University, S.P road, Guindy, Chennai, Tamil Nadu 600025, India
| | - Subash Babu
- NIH-TRC-ICER SAIC-Frederick Inc, National Cancer Institute at Frederick, Chetpet, Chennai, India
| | | |
Collapse
|
48
|
Metenou S, Kovacs M, Dembele B, Coulibaly YI, Klion AD, Nutman TB. Interferon regulatory factor modulation underlies the bystander suppression of malaria antigen-driven IL-12 and IFN-γ in filaria-malaria co-infection. Eur J Immunol 2012; 42:641-50. [PMID: 22213332 PMCID: PMC3430845 DOI: 10.1002/eji.201141991] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 11/09/2011] [Accepted: 12/02/2011] [Indexed: 01/12/2023]
Abstract
In areas where polyparasitism is highly prevalent, the impact of multiple parasites on the host response is underestimated. In particular, the presence of helminth infection coincident with malaria profoundly alters the production of malaria-specific IFN-γ, IL-12p70, CXCL9, CXCL10 and CXCL11, cytokines/chemokines known to be critical in mediating malaria-specific immunity. In order to elucidate the mechanisms underlying the suppression of malaria-specific cytokines/chemokines, we assessed the expression of malaria-specific IL-12Rβ1, IL-12Rβ2 and interferon regulatory factor (IRF)-1 in blood obtained from 18 filaria-infected (Fil(+)) and 17 filaria-uninfected (Fil(-)) individuals in a filaria-malaria co-endemic region of Mali. We found that Fil(+) individuals had significantly lower RNA expression of IRF-1 but not IL-12Rβ1 or IL-12Rβ2 in response to malaria antigen stimulation. We also measured the frequency of IL-12-producing DCs from these subjects and found that Fil(+) subjects had lower frequencies of IL-12(+) mDCs after malaria antigen stimulation than did the Fil(-) subjects. Modeling these data in vitro, we found that mDCs pre-exposed to live microfilariae not only produced significantly lower levels of CXCL-9, CXCL-10, IL-12p35, IL-12p40, IL-12p19 and CXCL-11 following stimulation with malaria antigen but also markedly downregulated the expression of IRF-1, IRF-2 and IRF-3 compared with microfilaria-unexposed mDCs. siRNA-inhibition of irf-1 in mDCs downregulated the production of IL-12p70 through repression of IL-12p35. Our data demonstrate that the modulation of IRFs seen in filarial (and presumably other tissue-invasive helminths) infection underlies the suppression of malaria-specific cytokines/chemokines that play a crucial role in immunity to malaria.
Collapse
Affiliation(s)
- Simon Metenou
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Steel C, Varma S, Nutman TB. Regulation of global gene expression in human Loa loa infection is a function of chronicity. PLoS Negl Trop Dis 2012; 6:e1527. [PMID: 22389737 PMCID: PMC3289604 DOI: 10.1371/journal.pntd.0001527] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 01/01/2012] [Indexed: 01/22/2023] Open
Abstract
Background Human filarial infection is characterized by downregulated parasite-antigen specific T cell responses but distinct differences exist between patients with longstanding infection (endemics) and those who acquired infection through temporary residency or visits to filarial-endemic regions (expatriates). Methods and Findings To characterize mechanisms underlying differences in T cells, analysis of global gene expression using human spotted microarrays was conducted on CD4+ and CD8+ T cells from microfilaremic Loa loa-infected endemic and expatriate patients. Assessment of unstimulated cells showed overexpression of genes linked to inflammation and caspase-associated cell death, particularly in endemics, and enrichment of the Th1/Th2 canonical pathway in endemic CD4+ cells. However, pathways within CD8+ unstimulated cells were most significantly enriched in both patient groups. Antigen (Ag)-driven gene expression was assessed to microfilarial Ag (MfAg) and to the nonparasite Ag streptolysin O (SLO). For MfAg-driven cells, the number of genes differing significantly from unstimulated cells was greater in endemics compared to expatriates (p<0.0001). Functional analysis showed a differential increase in genes associated with NFkB (both groups) and caspase activation (endemics). While the expatriate response to MfAg was primarily a CD4+ pro-inflammatory one, the endemic response included CD4+ and CD8+ cells and was linked to insulin signaling, histone complexes, and ubiquitination. Unlike the enrichment of canonical pathways in CD8+ unstimulated cells, both groups showed pathway enrichment in CD4+ cells to MfAg. Contrasting with the divergent responses to MfAg seen between endemics and expatriates, the CD4+ response to SLO was similar; however, CD8+ cells differed strongly in the nature and numbers (156 [endemics] vs 36 [expatriates]) of genes with differential expression. Conclusions These data suggest several important pathways are responsible for the different outcomes seen among filarial-infected patients with varying levels of chronicity and imply an important role for CD8+ cells in some of the global changes seen with lifelong exposure. Infection with the filarial parasite Loa loa causes a parasite-specific downregulation of T cell responses. However, differences exist (clinical and immunologic) between patients born and living in filarial endemic regions (endemics) and those who become infected during travel or short-term residency (expatriates). T cell responses are more depressed in endemics while expatriates have more clinical “allergic-type” symptoms. In this study, we showed that these differences reflect transcriptional differences within the T cell compartment. Using microarrays, we examined global gene expression in both CD4+ and CD8+ T cells of microfilaremic endemic and expatriate patients and found differences not only ex vivo, but also to parasite and, for CD8+ cells, to nonparasite antigens. Functional analysis showed that endemic patients expressed genes linked to inflammatory disease and caspase associated cell death at homeostasis while expatriates tended to have a more activation-induced gene profile at homeostasis and a CD4+ inflammatory response to parasite antigen. Patient groups were similar in their CD4+ response to nonparasite antigen but strongly differed in their CD8+ responses, demonstrating the potential global ramifications of chronic, longstanding infection. Our study describes potential transcriptional mechanisms for the variability seen in patients with different levels of exposure to and chronicity of filarial infection.
Collapse
Affiliation(s)
- Cathy Steel
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | |
Collapse
|
50
|
Nono JK, Pletinckx K, Lutz MB, Brehm K. Excretory/secretory-products of Echinococcus multilocularis larvae induce apoptosis and tolerogenic properties in dendritic cells in vitro. PLoS Negl Trop Dis 2012; 6:e1516. [PMID: 22363826 PMCID: PMC3283565 DOI: 10.1371/journal.pntd.0001516] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 12/19/2011] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Alveolar echinococcosis, caused by Echinococcus multilocularis larvae, is a chronic disease associated with considerable modulation of the host immune response. Dendritic cells (DC) are key effectors in shaping the immune response and among the first cells encountered by the parasite during an infection. Although it is assumed that E.multilocularis, by excretory/secretory (E/S)-products, specifically affects DC to deviate immune responses, little information is available on the molecular nature of respective E/S-products and their mode of action. METHODOLOGY/PRINCIPAL FINDINGS We established cultivation systems for exposing DC to live material from early (oncosphere), chronic (metacestode) and late (protoscolex) infectious stages. When co-incubated with Echinococcus primary cells, representing the invading oncosphere, or metacestode vesicles, a significant proportion of DC underwent apoptosis and the surviving DC failed to mature. In contrast, DC exposed to protoscoleces upregulated maturation markers and did not undergo apoptosis. After pre-incubation with primary cells and metacestode vesicles, DC showed a strongly impaired ability to be activated by the TLR ligand LPS, which was not observed in DC pre-treated with protoscolex E/S-products. While none of the larvae induced the secretion of pro-inflammatory IL-12p70, the production of immunosuppressive IL-10 was elevated in response to primary cell E/S-products. Finally, upon incubation with DC and naïve T-cells, E/S-products from metacestode vesicles led to a significant expansion of Foxp3+ T cells in vitro. CONCLUSIONS This is the first report on the induction of apoptosis in DC by cestode E/S-products. Our data indicate that the early infective stage of E. multilocularis is a strong inducer of tolerance in DC, which is most probably important for generating an immunosuppressive environment at an infection phase in which the parasite is highly vulnerable to host attacks. The induction of CD4+CD25+Foxp3+ T cells through metacestode E/S-products suggests that these cells fulfill an important role for parasite persistence during chronic echinococcosis.
Collapse
Affiliation(s)
- Justin Komguep Nono
- University of Würzburg, Institute of Hygiene and Microbiology, Würzburg, Germany
| | - Katrien Pletinckx
- University of Würzburg, Institute of Virology and Immunobiology, Würzburg, Germany
| | - Manfred B. Lutz
- University of Würzburg, Institute of Virology and Immunobiology, Würzburg, Germany
| | - Klaus Brehm
- University of Würzburg, Institute of Hygiene and Microbiology, Würzburg, Germany
| |
Collapse
|