1
|
Yu L, Li Q, Jiang N, Fan R, Zhang N, Zhang Y, Sun W, Chen R, Feng Y, Sang X, Chen Q. Toll-like receptor 9 signaling is associated with immune responses to Trypanosoma brucei infection. Int Immunopharmacol 2024; 134:112250. [PMID: 38749335 DOI: 10.1016/j.intimp.2024.112250] [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: 04/13/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 06/03/2024]
Abstract
Trypanosoma brucei, a causative agent of human and animal trypanosomiasis, regularly switches its major surface antigen to avoid elimination by the immune system. Toll-like receptor 9 (TLR9) is a key modulator for resistance to host-infective trypanosomes; however, the underlying molecular mechanism remains indistinct. Thus, we first approached the issue using Tlr9-mutant mice that render them non-responsive to TLR9 agonists. After infection, T cells in the spleens of Tlr9-mutant mice were analyzed by flow cytometry and a reduction in CD8+, CD4+ T, and NKT cells was observed in Tlr9-mutant mice compared to WT mice. We further found that the responses of inflammatory cytokines in the sera were reduced in Tlr9-mutant mice after T. brucei infection. The underlying molecular mechanism was that T. b. brucei DNA activated TLR9, which consequently upregulated the expression of p38 and ERK/MAPK, resulting in host resistance to trypanosome infection. In conclusion, these findings provide novel insights into the TLR9-mediated host responses to trypanosome infection.
Collapse
Affiliation(s)
- Liying Yu
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Qilong Li
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Ruiming Fan
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Naiwen Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Yiwei Zhang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Weisong Sun
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Ran Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Ying Feng
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Xiaoyu Sang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, and Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China.
| |
Collapse
|
2
|
Gheibi-Hayat SM, Jamialahmadi K. Antisense Oligonucleotide (AS-ODN) Technology: Principle, Mechanism and Challenges. Biotechnol Appl Biochem 2020; 68:1086-1094. [PMID: 32964539 DOI: 10.1002/bab.2028] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/12/2020] [Indexed: 12/20/2022]
Abstract
Recently, there is a hopefully tremendous interest in antisense therapeutics for clinical purposes. Single-stranded synthetic antisense oligonucleotides (As-ODNs) with monomers of chemically modified 18-21 deoxynucleotides complement the mRNA sequence in target gene. The target gene expression can be blocked because of created cleavage or disability of the mRNA by binding the As-ODNs to cognate mRNA sequences via sequence-specific hybridization. The idea of antisense therapy has become particular concerning that any sequence longer than a minimal number of nucleotides (17 for DNA and 13 for RNA) can be observed only once within the human genome. The mRNA is omnipresent more probably to manipulate compared to DNA, which results in multiple in vitro and in vivo applications for As-ODNs in the field of regulatory mechanisms of biological processes, cancer, viral infections and hereditary impairments. Although, there are uncertain clinical outcomes on the ability of this approach in treatment procedures despite achieving promising findings based on previous investigations. Accordingly, the efficacy, off-target effects, delivery are issues that should be investigated to obtain satisfactory results. In this review, we will explain the mechanism of action of As-ODNs and various types of modifications and their therapeutic purposes. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
Radwanska M, Vereecke N, Deleeuw V, Pinto J, Magez S. Salivarian Trypanosomosis: A Review of Parasites Involved, Their Global Distribution and Their Interaction With the Innate and Adaptive Mammalian Host Immune System. Front Immunol 2018; 9:2253. [PMID: 30333827 PMCID: PMC6175991 DOI: 10.3389/fimmu.2018.02253] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/11/2018] [Indexed: 01/27/2023] Open
Abstract
Salivarian trypanosomes are single cell extracellular parasites that cause infections in a wide range of hosts. Most pathogenic infections worldwide are caused by one of four major species of trypanosomes including (i) Trypanosoma brucei and the human infective subspecies T. b. gambiense and T. b. rhodesiense, (ii) Trypanosoma evansi and T. equiperdum, (iii) Trypanosoma congolense and (iv) Trypanosoma vivax. Infections with these parasites are marked by excessive immune dysfunction and immunopathology, both related to prolonged inflammatory host immune responses. Here we review the classification and global distribution of these parasites, highlight the adaptation of human infective trypanosomes that allow them to survive innate defense molecules unique to man, gorilla, and baboon serum and refer to the discovery of sexual reproduction of trypanosomes in the tsetse vector. With respect to the immunology of mammalian host-parasite interactions, the review highlights recent findings with respect to the B cell destruction capacity of trypanosomes and the role of T cells in the governance of infection control. Understanding infection-associated dysfunction and regulation of both these immune compartments is crucial to explain the continued failures of anti-trypanosome vaccine developments as well as the lack of any field-applicable vaccine based anti-trypanosomosis intervention strategy. Finally, the link between infection-associated inflammation and trypanosomosis induced anemia is covered in the context of both livestock and human infections.
Collapse
Affiliation(s)
- Magdalena Radwanska
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea
| | - Nick Vereecke
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Violette Deleeuw
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Joar Pinto
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Stefan Magez
- Laboratory for Biomedical Research, Ghent University Global Campus, Incheon, South Korea.,Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
4
|
López-Muñoz RA, Molina-Berríos A, Campos-Estrada C, Abarca-Sanhueza P, Urrutia-Llancaqueo L, Peña-Espinoza M, Maya JD. Inflammatory and Pro-resolving Lipids in Trypanosomatid Infections: A Key to Understanding Parasite Control. Front Microbiol 2018; 9:1961. [PMID: 30186271 PMCID: PMC6113562 DOI: 10.3389/fmicb.2018.01961] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 08/02/2018] [Indexed: 12/30/2022] Open
Abstract
Pathogenic trypanosomatids (Trypanosoma cruzi, Trypanosoma brucei, and Leishmania spp.) are protozoan parasites that cause neglected diseases affecting millions of people in Africa, Asia, and the Americas. In the process of infection, trypanosomatids evade and survive the immune system attack, which can lead to a chronic inflammatory state that induces cumulative damage, often killing the host in the long term. The immune mediators involved in this process are not entirely understood. Most of the research on the immunologic control of protozoan infections has been focused on acute inflammation. Nevertheless, when this process is not terminated adequately, permanent damage to the inflamed tissue may ensue. Recently, a second process, called resolution of inflammation, has been proposed to be a pivotal process in the control of parasite burden and establishment of chronic infection. Resolution of inflammation is an active process that promotes the normal function of injured or infected tissues. Several mediators are involved in this process, including eicosanoid-derived lipids, cytokines such as transforming growth factor (TGF)-β and interleukin (IL)-10, and other proteins such as Annexin-V. For example, during T. cruzi infection, pro-resolving lipids such as 15-epi-lipoxin-A4 and Resolvin D1 have been associated with a decrease in the inflammatory changes observed in experimental chronic heart disease, reducing inflammation and fibrosis, and increasing host survival. Furthermore, Resolvin D1 modulates the immune response in cells of patients with Chagas disease. In Leishmania spp. infections, pro-resolving mediators such as Annexin-V, lipoxins, and Resolvin D1 are related to the modulation of cutaneous manifestation of the disease. However, these mediators seem to have different roles in visceral or cutaneous leishmaniasis. Finally, although T. brucei infections are less well studied in terms of their relationship with inflammation, it has been found that arachidonic acid-derived lipids act as key regulators of the host immune response and parasite burden. Also, cytokines such as IL-10 and TGF-β may be related to increased infection. Knowledge about the inflammation resolution process is necessary to understand the host–parasite interplay, but it also offers an interesting opportunity to improve the current therapies, aiming to reduce the detrimental state induced by chronic protozoan infections.
Collapse
Affiliation(s)
- Rodrigo A López-Muñoz
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Alfredo Molina-Berríos
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Carolina Campos-Estrada
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso, Chile.,Centro de Investigación Farmacopea Chilena, Universidad de Valparaíso, Valparaíso, Chile
| | - Patricio Abarca-Sanhueza
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Luis Urrutia-Llancaqueo
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Miguel Peña-Espinoza
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Juan D Maya
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
5
|
Masocha W, Kristensson K. Human African trypanosomiasis: How do the parasites enter and cause dysfunctions of the nervous system in murine models? Brain Res Bull 2018; 145:18-29. [PMID: 29870779 DOI: 10.1016/j.brainresbull.2018.05.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/24/2018] [Accepted: 05/30/2018] [Indexed: 12/27/2022]
Abstract
In this review we describe how Trypanosoma brucei brucei, a rodent pathogenic strain of African trypanosomes, can invade the nervous system, first by localization to the choroid plexus, the circumventricular organs (CVOs) and peripheral ganglia, which have fenestrated vessels, followed by crossing of the blood-brain barrier (BBB) into the white matter, hypothalamus, thalamus and basal ganglia. White blood cells (WBCs) pave the way for the trypanosome neuroinvasion. Experiments with immune deficient mice show that the invasion of WBCs is initiated by the toll-like receptor 9, followed by an augmentation phase that depends on the cytokine IFN-γ and the chemokine CXCL10. Nitric oxide (NO) derived from iNOS then prevents a break-down of the BBB and non-regulated passage of cells. This chain of events is relevant for design of better diagnostic tools to distinguish the different stages of the disease as well as for better understanding of the pathogenesis of the nervous system dysfunctions, which include circadian rhythm changes with sleep pattern disruption, pain syndromes, movement disorders and mental disturbances including dementia.
Collapse
Affiliation(s)
- Willias Masocha
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Kuwait University, Kuwait.
| | | |
Collapse
|
6
|
Zhang CX, Chen J, Cai L, Wu J, Wang JY, Cao LF, Zhou W, Chen TX. DNA induction of MDM2 promotes proliferation of human renal mesangial cells and alters peripheral B cells subsets in pediatric systemic lupus erythematosus. Mol Immunol 2018; 94:166-175. [PMID: 29324237 DOI: 10.1016/j.molimm.2018.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 12/11/2017] [Accepted: 01/04/2018] [Indexed: 02/06/2023]
Abstract
The study is aimed to investigate the role of MDM2 in the pathogenesis of lupus nephritis (LN) in pediatric SLE (pSLE). We confirmed that MDM2 expression was increased in peripheral blood mononuclear cells (PBMCs) as well as renal specimen of SLE compared with that of controls by western blot and immunofluorescence staining. Percentage of apoptotic and necrotic CD4+T, CD8+T and B cells were detected by flow cytometry respectively and levels of plasma cell free DNA (cfDNA) were quantified in SLE and healthy controls (HC). We also proved that elevated apoptotic and necrotic CD4+T cells were the main cause for increased plasma levels of cfDNA in pSLE. Additionally, upon DNA transfection MDM2 increased while P53 and P21 decreased in human renal mesangial cells (HRMC), with concomitant increase in proliferation rate and proportion of cells in S phase, as demonstrated by cell proliferation assay and cell cycle analysis. However, MDM2 inhibition reversed the trend. Furthermore, percentage of switched memory B cells decreased and proportion of double negative B cells increased upon blockage of MDM2 in PBMC. In summary, our study provided the first evidence that DNA induction of MDM2 promotes proliferation of HRMC and alters peripheral B cells subsets in pSLE. Thus our study has not only elucidated the pathogenesis of MDM2 in pediatric LN but also provided a novel target for drug development. In conclusion, our data suggested that apoptosis, cfDNA and MDM2 could form a pathological axis in SLE, especially in pSLE.
Collapse
Affiliation(s)
- Chen-Xing Zhang
- Department of Allergy and Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Ji Chen
- Department of Dermatology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Li Cai
- Department of Allergy and Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Jing Wu
- Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Jia-Yuan Wang
- Department of Laboratory Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Lan-Fang Cao
- Department of Pediatrics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Wei Zhou
- Department of Nephrology and Rheumatology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Tong-Xin Chen
- Department of Allergy and Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Department of Nephrology and Rheumatology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; Division of Immunology, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
7
|
Black SJ, Mansfield JM. Prospects for vaccination against pathogenic African trypanosomes. Parasite Immunol 2017; 38:735-743. [PMID: 27636100 DOI: 10.1111/pim.12387] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 09/14/2016] [Indexed: 11/27/2022]
Abstract
African trypanosomes cause human and animal African trypanosomiases, which are chronic, debilitating and often fatal diseases of people and livestock in sub-Saharan Africa. The extracellular protozoan parasites are exemplars of antigenic variation. They direct host-protective B-cell and T-cell immune responses towards hypervariable components of their variable surface glycoprotein coat and evade immune elimination by generating new surface coat antigenic variants at a rate that supersedes immune destruction. This results in recurring waves of parasitemia, tissue invasion and escalating immunopathology in trypanosomiasis-susceptible hosts. Here, we discuss the possibility that host control of African trypanosomes might be improved by immunization with conserved VSG peptides and invariant surface glycoproteins. Infection-induced T-cell recall responses to these typically poorly expressed or nonimmunogenic parasite components induce tissue phagocytes to produce microbicidal materials that kill trypanosomes. Preliminary data that support this immune-enhancing vaccine strategy are discussed, as are host and parasite interactions that might downregulate the protective responses. These include infection-induced immunosuppression and increasing virulence of infecting parasites over time.
Collapse
Affiliation(s)
- S J Black
- Department of Veterinary and Animal Sciences, University of Massachusetts-Amherst, Amherst, MA, USA
| | - J M Mansfield
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
8
|
Dos-Santos A, Carvalho-Kelly L, Dick C, Meyer-Fernandes J. Innate immunomodulation to trypanosomatid parasite infections. Exp Parasitol 2016; 167:67-75. [DOI: 10.1016/j.exppara.2016.05.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 05/13/2016] [Accepted: 05/21/2016] [Indexed: 01/05/2023]
|
9
|
Zhao F, Li YW, Pan HJ, Shi CB, Luo XC, Li AX, Wu SQ. Expression profiles of toll-like receptors in channel catfish (Ictalurus punctatus) after infection with Ichthyophthirius multifiliis. FISH & SHELLFISH IMMUNOLOGY 2013; 35:993-997. [PMID: 23742868 DOI: 10.1016/j.fsi.2013.05.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 05/10/2013] [Accepted: 05/20/2013] [Indexed: 06/02/2023]
Abstract
Toll-like receptors (TLRs) play a crucial role in the innate immune system, but to date the roles of fish TLRs in response to parasitic infection are still poorly understood. In the present study, we used channel catfish (Ictalurus punctatus) and the ciliate parasite Ichthyophthirius multifiliis as a model to investigate whether and which fish TLRs play important roles in the immune response against parasitic pathogens by detecting the expression profiles of a complete set of TLRs in catfish at different time points after infection with I. multifiliis. The expression profiles of TLR1 and TLR2 were similar, and both were significantly up-regulated in the skin and head kidney at most time points after infection. Furthermore, the expression of TLR2 was also up-regulated in the gill and spleen. TLR9 was induced in the skin and gill, whereas TLR21 was induced in the head kidney and spleen after infection. For TLR19, significant up-regulation was observed in the skin and gill, but significant down-regulation was detected in the head kidney and spleen. In contrast to TLR19, TLR25 was significantly up-regulated in the head kidney and spleen at some time points. No significant changes were observed for the rest of the TLRs at most time points. The results indicated that some TLRs may play essential roles in catfish defense against I. multifiliis infection.
Collapse
Affiliation(s)
- Fei Zhao
- Key Laboratory of Fishery Drug Development, Ministry of Agriculture, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, 1 Xingyu Road, Liwan District, Guangzhou 510380, Guangdong Province, PR China
| | | | | | | | | | | | | |
Collapse
|
10
|
Al-Mathkhury HJF, Abdul-Ghaffar SN. Urinary tract infections caused by staphylococcus aureus DNA in comparison to the candida albicans DNA. NORTH AMERICAN JOURNAL OF MEDICAL SCIENCES 2012; 3:565-9. [PMID: 22363080 PMCID: PMC3271421 DOI: 10.4297/najms.2011.3562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Background: Bacterial DNA released upon bacterial autolysis or killed by antibiotics, hence, many inflammatogenic reactions will be established leading to serious tissue damage. Aim: the present work aimed to elucidate the histopathological changes caused by prokaryotic (bacterial) DNA and eukaryotic (candidal) DNA. Materials and Methods: twenty one Staphylococcus aureus and 36 Candida albicans isolates were isolated from UTI patients. Viable cells and DNA of the highest antibiotic sensitive isolates were injected, intraurethraly, in mice. Results were evaluated via histopathological examination. Results: Mildest reactions were obtained from mice challenged with viable C. albicans compared with those challenged with viable S. aureus. Dose-dependent histological changes were observed for both eukaryotic and prokaryotic DNA. However, the eukaryotic C. albicans DNA developed less intense histological changes than S. aureus DNA. Conclusion: microbial DNA has the ability to cause damage in murine renal system. Nevertheless, bacterial DNA caused more intense damage than candidal DNA.
Collapse
|
11
|
Host cell signalling and leishmania mechanisms of evasion. J Trop Med 2011; 2012:819512. [PMID: 22131998 PMCID: PMC3216306 DOI: 10.1155/2012/819512] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 08/16/2011] [Indexed: 12/18/2022] Open
Abstract
Leishmania parasites are able to secure their survival and propagation within their host by altering signalling pathways involved in the ability of macrophages to kill pathogens or to engage adaptive immune system. An important step in this immune evasion process is the activation of host protein tyrosine phosphatase SHP-1 by Leishmania. SHP-1 has been shown to directly inactivate JAK2 and Erk1/2 and to play a role in the negative regulation of several transcription factors involved in macrophage activation. These signalling alterations contribute to the inactivation of critical macrophage functions (e.g., Nitric oxide, IL-12, and TNF-α). Additionally, to interfere with IFN-γ receptor signalling, Leishmania also alters several LPS-mediated responses. Recent findings from our laboratory revealed a pivotal role for SHP-1 in the inhibition of TLR-induced macrophage activation through binding to and inactivating IL-1-receptor-associated kinase 1 (IRAK-1). Furthermore, we identified the binding site as an evolutionarily conserved ITIM-like motif, which we named kinase tyrosine-based inhibitory motif (KTIM). Collectively, a better understanding of the evasion mechanisms utilized by Leishmania parasite could help to develop more efficient antileishmanial therapies in the near future.
Collapse
|
12
|
Abstract
The experimental studies of Brucei group trypanosomes presented here demonstrate that the balance of host and parasite factors, especially IFN-γ GPI-sVSG respectively, and the timing of cellular exposure to them, dictate the predominant MP and DC activation profiles present at any given time during infection and within specific tissues. The timing of changes in innate immune cell functions following infection consistently support the conclusion that the key events controlling host resistance occur within a short time following initial exposure to the parasite GPI substituents. Once the changes in MP and DC activities are initiated, there appears little that the host can do to reverse these changes and alter the final outcome of these regulatory events. Instead, despite the availability of multiple innate and adaptive immune mechanisms that can control parasites, there is an inability to control trypanosome numbers sufficiently to prevent the emergence and establishment of virulent trypanosomes that eventually kill the host. Overall it appears that trypanosomes have carefully orchestrated the host innate and adaptive immune response so that parasite survival and transmission, and alterations of host immunity, are to its ultimate benefit.
Collapse
|
13
|
Khandwekar AP, Patil DP, Shouche Y, Doble M. Surface Engineering of Polycaprolactone by Biomacromolecules and their Blood Compatibility. J Biomater Appl 2010; 26:227-52. [DOI: 10.1177/0885328210367442] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Improving blood compatibility of biodegradable polymers is an area of intensive research in blood contacting devices. In this study, curdlan sulphate and heparin-modified poly (caprolactone) (PCL) hybrids were developed by physically entrapping these molecules on the PCL surface. This modification technique was performed by reversible gelation of the PCL surface region following exposure to a solvent and nonsolvent mixture. The presence of these biomacromolecules on the PCL surface was verified by atomic force microscopy (AFM) and scanning electron microscopy and energy dispersive X-ray analysis (SEM-EDAX) analysis, while wettability of the films was investigated by dynamic contact angle measurements. The blood compatibilities of the surface-modified films were examined using in vitro platelet and leukocyte adhesion and thrombus formation. Mouse RAW 264.7 macrophage cells were used to assess the cell adhesion and inflammatory response to the modified surface by quantifying mRNA expression levels of proinflammatory cytokines namely TNF-α and IL-1β using real-time polymerase chain reaction (RT-PCR). A lower platelet and leukocyte adhesion and activation was observed on the modified films incubated with whole human blood for 2 h. The thrombus formation on the PCL was significantly decreased upon immobilization of both curdlan sulphate (39%, *p<0.05) and heparin (28%, *p<0.01) when compared to bare PCL (80%). All of these results revealed that improved blood compatibility was obtained by surface entrapment of both curdlan sulphate (CURS) and heparin (HEP) onto PCL films. Both PCL-CURS and PCL-HEP films reduced RAW 264.7 macrophage cell adhesion (*p<0.05) with respect to the base unmodified PCL. The cellular inflammatory response was suppressed on the modified substrates. The mRNA expression levels of proinflammmatory cytokines (TNF-α and IL-1β) were upregulated on bare PCL, while it was significantly lower on PCL-CURS and PCL-HEP substrates (**p<0.001). Thus, this biomacromolecule entrapment process can be applied on PCL in order to achieve improved blood compatibility and reduced inflammatory host response for its future blood contacting applications.
Collapse
Affiliation(s)
- Anand P. Khandwekar
- Department of Biotechnology, Indian Institute of Technology Madras Chennai 600036, India
| | - Deepak P. Patil
- National Center for Cell Science, Ganeshkhind, Pune 411007, India
| | - Yogesh Shouche
- National Center for Cell Science, Ganeshkhind, Pune 411007, India
| | - Mukesh Doble
- Department of Biotechnology, Indian Institute of Technology Madras Chennai 600036, India,
| |
Collapse
|
14
|
Immunobiology of African trypanosomes: need of alternative interventions. J Biomed Biotechnol 2010; 2010:389153. [PMID: 20182644 PMCID: PMC2826769 DOI: 10.1155/2010/389153] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 10/29/2009] [Accepted: 12/23/2009] [Indexed: 02/08/2023] Open
Abstract
Trypanosomiasis is one of the major parasitic diseases for which control is still far from reality. The vaccination approaches by using dominant surface proteins have not been successful, mainly due to antigenic variation of the parasite surface coat. On the other hand, the chemotherapeutic drugs in current use for the treatment of this disease are toxic and problems of resistance are increasing (see Kennedy (2004) and Legros et al. (2002)). Therefore, alternative approaches in both treatment and vaccination against trypanosomiasis are needed at this time. To be able to design and develop such alternatives, the biology of this parasite and the host response against the pathogen need to be studied. These two aspects of this disease with few examples of alternative approaches are discussed here.
Collapse
|
15
|
Khandwekar AP, Patil DP, Shouche YS, Doble M. The biocompatibility of sulfobetaine engineered polymethylmethacrylate by surface entrapment technique. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2010; 21:635-646. [PMID: 19821070 DOI: 10.1007/s10856-009-3886-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Accepted: 09/30/2009] [Indexed: 05/28/2023]
Abstract
Sulfobetaine-modified polymethylmethacrylate (PMMA) systems were created by physically entrapping the zwitterionic species on the PMMA surface. The presence of the sulfobetaine molecules on these surfaces were verified by ATR-FTIR and SEM-EDAX analysis, while wettability of the films was investigated by dynamic contact angle measurements. The short-term (4 h) adhesion of two bacterial species (gram-positive Staphylococcus aureus and gram-negative Pseudomonas aeruginosa) on these surfaces were studied. Mouse RAW 264.7 macrophage cells were used to assess the cell adhesion and inflammatory response by quantifying the expression levels of proinflammatory cytokines namely TNFalpha and IL1beta by measuring their mRNA profiles in the cells using real-time polymerase chain reaction (RT-PCR) normalized to the house keeping gene GAPDH. Whilst mouse L-929 fibroblast cells were used to assess the propensity for the materials to support fibroblast cell adhesion. A decrease in the adhesion of S. aureus by 63% and P. aeruginosa by 49% was observed on sulfobetaine modified PMMA films after 4 h. In all the cases, sulfobetaine modified PMMA films reduced cellular adhesion events (*P < 0.05) with respect to the base materials, which could be linked to the reduced protein adsorption observed on these surfaces. The cellular inflammatory response was suppressed on sulfobetaine modified substrates as expression levels of pro-inflammatory cytokines (TNFalpha and IL1beta) was found to be up regulated on bare PMMA, while it was significantly lower on sulfobetaine modified PMMA (**P < 0.001). Thus the sulfobetaine entrapment process can be applied on polymethylmethacrylate in order to achieve low biointeractions and reduced inflammatory host responses for various biomedical and biotechnological applications.
Collapse
Affiliation(s)
- Anand P Khandwekar
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, 600036, India.
| | | | | | | |
Collapse
|
16
|
Das UN. Do long-chain unsaturated fatty acids function as endogenous anti-trypanosomal molecules? Med Hypotheses 2009; 74:676-8. [PMID: 19962250 DOI: 10.1016/j.mehy.2009.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2009] [Revised: 11/03/2009] [Accepted: 11/04/2009] [Indexed: 11/15/2022]
Abstract
Trypanosomiasis is common in Africa and South America. The surface of trypanosomes is covered by glycosyl phosphatidylinositol (GPI)-anchored variant surface glycoprotein (VSG) that contains the fatty acid myristate (14:0) as its lipid moiety that is essential for its survival. Myristic acid, being a saturated fatty acid, is resistant to peroxidation. Hence, I propose that replacement of myristic acid of VSG-GPI of trypanosomes by unsaturated fatty acids may render them unviable due to the ease with which the latter undergo peroxidation. In addition, unsaturated fatty acids could kill trypanosomes by (1) activation of macrophages and release of reactive oxygen species; (2) high Ca(2+) influx that may be lethal; (3) alteration in mitochondrial membrane potential and disruption of energy generation; and (4) direct disruption of the cell membrane. If this hypothesis proves to be correct, unsaturated fatty acids and its analogues could form a new approach to the management of trypanosomiasis and other intracellular parasitic infections.
Collapse
Affiliation(s)
- Undurti N Das
- UND Life Sciences, 13800 Fairhill Road, #321, Shaker Heights, OH 44120, USA
| |
Collapse
|
17
|
Dagenais TR, Freeman BE, Demick KP, Paulnock DM, Mansfield JM. Processing and presentation of variant surface glycoprotein molecules to T cells in African trypanosomiasis. THE JOURNAL OF IMMUNOLOGY 2009; 183:3344-55. [PMID: 19675169 DOI: 10.4049/jimmunol.0802005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Th1 cell responses to the variant surface glycoprotein (VSG) of African trypanosomes play a critical role in controlling infection through the production of IFN-gamma, but the role of APCs in the induction and regulation of T cell-mediated protection is poorly understood. In this study, we have investigated the Ag presentation capabilities of dendritic cells (DCs) and macrophages during early trypanosome infection in relatively resistant responder and susceptible nonresponder mouse strains. Splenic DCs appeared to be the primary cell responsible for activating naive VSG-specific Th cell responses in resistant responder animals through the coordinated up-regulation of costimulatory molecules, secretion of IL-12, and presentation of VSG peptides to T cells in vivo. Splenic DC depletion and the down-regulation of costimulatory markers on splenic macrophages were observed in susceptible animals and may be associated with the inability of these animals to elicit a significant VSG-specific T cell response. In contrast to splenic APCs, peritoneal macrophages secreted NO, failed to activate naive Th cells in vitro, and presented relatively low levels of VSG peptides to T cells in vivo. Thus, VSG-specific Th1 cell responses may be determined by tissue- and cell-specific differences in Ag presentation. Additionally, all APCs from resistant and susceptible strains displayed a reduced ability to process and present newly encountered exogenous Ag, including new VSG molecules, during high parasitemia. Thus, initial uptake of VSG (or other trypanosome factors) may interfere with Ag presentation and have dramatic consequences for subsequent T cell responses to other proteins.
Collapse
Affiliation(s)
- Taylor R Dagenais
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | | | | | |
Collapse
|
18
|
Bosschaerts T, Guilliams M, Stijlemans B, De Baetselier P, Beschin A. Understanding the role of monocytic cells in liver inflammation using parasite infection as a model. Immunobiology 2009; 214:737-47. [PMID: 19577324 DOI: 10.1016/j.imbio.2009.06.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Uncontrolled inflammation is a major cause of pathogenicity during chronic parasite infections. Novel therapies should therefore aim at re-establishing the balance between pro- and anti-inflammatory signals during disease to avoid tissue damage and ensure survival of the host. In this context, we are intending to identify strategies capable of inducing counter-inflammatory activity in injured liver and thereby increasing the resistance of the host to African trypanosomiasis as a model for parasite infection. Here, recent evidence is summarized revealing how monocytic cells recruited to the liver of African trypanosome-infected mice develop an M1 or M2 activation status, thereby maintaining the capacity of the host to control parasite growth while avoiding the development of liver damage, which otherwise culminates in early death of the host.
Collapse
Affiliation(s)
- Tom Bosschaerts
- Department of Molecular and Cellular Interactions, VIB, Brussel, Belgium
| | | | | | | | | |
Collapse
|
19
|
Khandwekar AP, Patil DP, Khandwekar V, Shouche YS, Sawant S, Doble M. TecoflexTM functionalization by curdlan and its effect on protein adsorption and bacterial and tissue cell adhesion. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2009; 20:1115-1129. [PMID: 19093193 DOI: 10.1007/s10856-008-3655-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Accepted: 11/24/2008] [Indexed: 05/27/2023]
Abstract
Curdlan modified polyurethane was created by physically entrapping the former on TecoflexTM surface. ATR-FT-IR, SEM-EDAX and AFM analysis revealed the formation of stable thin curdlan layer on the film. Contact-angle measurements showed that the modified film was highly hydrophilic. Confocal laser scanning microscopy showed the existence of entrapped layer of approximately 20-25 microm in depth. Surface entrapment of curdlan minimized both protein adsorption and mouse L929 fibroblast cell adhesion relative to the control. Surface induced cellular inflammatory response was determined from the expression levels of proinflammatory cytokine TNF-alpha, by measuring their mRNA profiles in the cells using real time polymerase chain reaction (RT-PCR) normalized to the housekeeping gene GAPDH. The inflammatory response was suppressed on the modified substrate as expression of TNF-alpha mRNA was found to be up regulated on TecoflexTM, while it was significantly lower on curdlan substrate. The adhesion of S. aureus decreased by 62% on curdlan modified surface. Using such simple surface entrapment process, it will be possible to develop well-defined surface modifications that promote specific cell interactions and perhaps better performance in the long-term as implant.
Collapse
Affiliation(s)
- Anand P Khandwekar
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
| | | | | | | | | | | |
Collapse
|
20
|
Egan CE, Sukhumavasi W, Butcher BA, Denkers EY. Functional aspects of Toll-like receptor/MyD88 signalling during protozoan infection: focus on Toxoplasma gondii. Clin Exp Immunol 2009; 156:17-24. [PMID: 19161444 DOI: 10.1111/j.1365-2249.2009.03876.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Toll-like receptor (TLR)/MyD88 signalling has emerged as a major pathway of pathogen recognition in the innate immune system. Here, we review recent data that begin to show how this pathway controls the immune response to protozoan infection, with particular emphasis on the opportunistic pathogen Toxoplasma gondii. The various ways that the parasite activates and suppresses TLR/MyD88 signalling defines several key principals that illuminate the complexities of the host-pathogen interaction. We also speculate how TLR/MyD88 signalling might be exploited to provide protection against Toxoplasma, as well as other protozoa and infection in general.
Collapse
Affiliation(s)
- C E Egan
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | | | | |
Collapse
|
21
|
Mishra BB, Gundra UM, Teale JM. Toll-like receptors in CNS parasitic infections. Curr Top Microbiol Immunol 2009; 336:83-104. [PMID: 19688329 DOI: 10.1007/978-3-642-00549-7_5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Parasite infections in the central nervous system (CNS) are a major cause of morbidity and mortality worldwide, second only to HIV infection. Finding appropriate therapeutic measures to control CNS parasite infections requires an understanding of the tissue-specific host response. CNS parasitic diseases are invariably associated with persistent T-helper 1 (Th1) cytokine-dependent proinflammatory responses. Although type 1 cytokine-dependent proinflammatory responses are essential to control several types of parasite infections, their persistent production contributes to the development of neuropathology with severe consequences. A family of proteins called Toll-like receptors (TLRs) plays a pivotal role in the induction of inflammatory cytokines during infections and tissue injury. Accumulating evidence indicates that in several CNS parasitic infections such as toxoplasmosis and sleeping sickness, host responses mediated through TLRs contribute to parasite clearance and host survival. However, TLR-mediated responses can also contribute to disease severity, as exemplified in cerebral malaria, neurocysticercosis and river blindness. Thus, TLRs influence the immunopathogenesis of CNS parasitic infections by mechanisms that can either benefit the host or further contribute to CNS pathology. This chapter discusses the immunopathogenesis of parasitic infections in the CNS and the role of TLRs in this process.
Collapse
Affiliation(s)
- Bibhuti B Mishra
- Department of Biology, South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249-1644, USA
| | | | | |
Collapse
|
22
|
Lopez R, Demick KP, Mansfield JM, Paulnock DM. Type I IFNs play a role in early resistance, but subsequent susceptibility, to the African trypanosomes. THE JOURNAL OF IMMUNOLOGY 2008; 181:4908-17. [PMID: 18802094 DOI: 10.4049/jimmunol.181.7.4908] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Macrophages express a spectrum of proinflammatory and regulatory mediators during African trypanosomiasis. Microarray analyses revealed similar profiles of induced genes in macrophages stimulated with the trypanosome soluble variant surface glycoprotein in vitro and in macrophages taken from infected mice. Genes associated with the acute phase response and with type I IFN responses were prominent components of the macrophage activation profiles expressed within 72 h in vitro and in vivo. Thus, induction of proinflammatory gene expression is a characteristic of early trypanosome infection that is driven primarily by soluble variant surface glycoprotein exposure, and it may be that IFN-alpha/beta plays a central role in regulation of early resistance to trypanosomes. To test this hypothesis, we assessed parameters of infection in mouse strains with genetic alterations in the IFN-alpha/beta response pathway. We found that Ifnar1(-/-) mice, which lack the receptor for type I IFNs, exhibited delayed control of parasite burden during the first week of infection and died earlier than did wild-type controls. However, infection of Ubp43(-/-) mice, which are hyperresponsive to type I IFNs, did not exhibit enhanced resistance to trypanosomes. Instead, these animals also failed to control parasite burden and were more susceptible than wild-type animals. Additionally, the Ubp43(-/-) mice exhibited a significant defect in IFN-gamma production, which is definitively linked to host resistance in trypanosomiasis. These results show that type I IFNs play a role in early control of parasites in infected mice but may contribute to down-regulation of IFN-gamma production and subsequent loss of host resistance later in infection.
Collapse
Affiliation(s)
- Rebecca Lopez
- Department of Medical Microbiology and Immunology, University of Wisconsin Medical School, Madison, WI 53706, USA
| | | | | | | |
Collapse
|
23
|
Bartholomeu DC, Ropert C, Melo MB, Parroche P, Junqueira CF, Teixeira SMR, Sirois C, Kasperkovitz P, Knetter CF, Lien E, Latz E, Golenbock DT, Gazzinelli RT. Recruitment and endo-lysosomal activation of TLR9 in dendritic cells infected with Trypanosoma cruzi. THE JOURNAL OF IMMUNOLOGY 2008; 181:1333-44. [PMID: 18606688 DOI: 10.4049/jimmunol.181.2.1333] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TLR9 is critical in parasite recognition and host resistance to experimental infection with Trypanosoma cruzi. However, no information is available regarding nucleotide sequences and cellular events involved on T. cruzi recognition by TLR9. In silico wide analysis associated with in vitro screening of synthetic oligonucleotides demonstrates that the retrotransposon VIPER elements and mucin-like glycoprotein (TcMUC) genes in the T. cruzi genome are highly enriched for CpG motifs that are immunostimulatory for mouse and human TLR9, respectively. Importantly, infection with T. cruzi triggers high levels of luciferase activity under NF-kappaB-dependent transcription in HEK cells cotransfected with human TLR9, but not in control (cotransfected with human MD2/TLR4) HEK cells. Further, we observed translocation of TLR9 to the lysosomes during invasion/uptake of T. cruzi parasites by dendritic cells. Consistently, potent proinflammatory activity was observed when highly unmethylated T. cruzi genomic DNA was delivered to the endo-lysosomal compartment of host cells expressing TLR9. Thus, together our results indicate that the unmethylated CpG motifs found in the T. cruzi genome are likely to be main parasite targets and probably become available to TLR9 when parasites are destroyed in the lysosome-fused vacuoles during parasite invasion/uptake by phagocytes.
Collapse
Affiliation(s)
- Daniella C Bartholomeu
- Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Prolactin and growth hormone induce differential cytokine and chemokine profile in murine peritoneal macrophages in vitro: Involvement of p-38 MAP kinase, STAT3 and NF-κB. Cytokine 2008; 41:162-73. [DOI: 10.1016/j.cyto.2007.11.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2007] [Revised: 10/29/2007] [Accepted: 11/23/2007] [Indexed: 11/20/2022]
|
25
|
Stijlemans B, Guilliams M, Raes G, Beschin A, Magez S, De Baetselier P. African trypanosomosis: From immune escape and immunopathology to immune intervention. Vet Parasitol 2007; 148:3-13. [PMID: 17560035 DOI: 10.1016/j.vetpar.2007.05.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
African trypanosomes can cause prolonged chronic infections through a mechanism of antigen variation whereby they manipulate the humoral immune system of their hosts. However, besides antigenic variation these extracellular parasites exert other immunoregulatory activities mainly mediated by innate cells in particular macrophage-like (M) cells. In this review, the modulation of M cells through parasite factors and host cytokines as well as their role in parasite control and immunopathology will be examined. The concept of M cell polarization into distinct activation states (M1, M2) that may contribute to trypanosusceptibility or resistance will be discussed. Finally, the possibility to interfere with such activation states hereby providing new therapeutical modalities in the treatment of this infectious disease will be illustrated.
Collapse
Affiliation(s)
- Benoît Stijlemans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium.
| | | | | | | | | | | |
Collapse
|
26
|
Charmoy M, Megnekou R, Allenbach C, Zweifel C, Perez C, Monnat K, Breton M, Ronet C, Launois P, Tacchini-Cottier F. Leishmania major induces distinct neutrophil phenotypes in mice that are resistant or susceptible to infection. J Leukoc Biol 2007; 82:288-99. [PMID: 17449725 DOI: 10.1189/jlb.0706440] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Polymorphonuclear neutrophils (PMN) are key components of the inflammatory response contributing to the development of pathogen-specific immune responses. Following infection with Leishmania major, neutrophils are recruited within hours to the site of parasite inoculation. C57BL/6 mice are resistant to infection, and BALB/c mice are susceptible to infection, developing unhealing, inflammatory lesions. In this report, we investigated the expression of cell surface integrins, TLRs, and the secretion of immunomodulatory cytokines by PMN of both strains of mice, in response to infection with L. major. The parasite was shown to induce CD49d expression in BALB/c-inflammatory PMN, and expression of CD49d remained at basal levels in C57BL/6 PMN. Equally high levels of CD11b were expressed on PMN from both strains. In response to L. major infection, the levels of TLR2, TLR7, and TLR9 mRNA were significantly higher in C57BL/6 than in BALB/c PMN. C57BL/6 PMN secreted biologically active IL-12p70 and IL-10. In contrast, L. major-infected BALB/c PMN transcribed and secreted high levels of IL-12p40 but did not secrete biologically active IL-12p70. Furthermore, IL-12p40 was shown not to associate with IL-23 p19 but formed IL-12p40 homodimers with inhibitory activity. No IL-10 was secreted by BALB/c PMN. Thus, following infection with L. major, in C57BL/6 mice, PMN could constitute one of the earliest sources of IL-12, and in BALB/c mice, secretion of IL-12p40 could contribute to impaired, early IL-12 signaling. These distinct PMN phenotypes may thus influence the development of L. major-specific immune response.
Collapse
Affiliation(s)
- Mélanie Charmoy
- WHO Immunology Research and Training Center, Department of Biochemistry, Epalinges, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Harris TH, Mansfield JM, Paulnock DM. CpG oligodeoxynucleotide treatment enhances innate resistance and acquired immunity to African trypanosomes. Infect Immun 2007; 75:2366-73. [PMID: 17339353 PMCID: PMC1865757 DOI: 10.1128/iai.01649-06] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Relative resistance to African trypanosomiasis is based on the development of a type I cytokine response, which is partially dependent on innate immune responses generated through MyD88 and Toll-like receptor 9 (TLR9). Therefore, we asked whether enhancement of the immune response by artificial stimulation with CpG oligodeoxynucleotide (ODN), a TLR9 agonist, would result in enhanced protection against trypanosomes. In susceptible BALB/c mice, relative resistance to infection was significantly enhanced by CpG ODN treatment and was associated with decreased parasite burden, increased cytokine production, and elevated parasite-specific B- and T-cell responses. In relatively resistant C57BL/6 mice, survival was not enhanced but early parasitemia levels were reduced 100-fold and the majority of the parasites were nondividing, short stumpy (SS) forms. CpG ODN treatment of lymphocyte-deficient C57BL/6-scid and BALB/cByJ-scid mice also enhanced survival and reduced parasitemia, indicating that innate resistance to trypanosome infection can be enhanced. In C57BL/6-scid and BALB/cByJ-scid mice, the parasites were also predominantly SS forms during the outgrowth of parasitemia. However, the effect of CpG ODN treatment on parasite morphology was not as marked in gamma interferon (IFN-gamma)-knockout mice, suggesting that downstream effects of IFN-gamma production may play a discrete role in parasite cell differentiation. Overall, these studies provide the first evidence that enhancement of resistance to African trypanosomes can be induced in susceptible animals in a TLR9-dependent manner and that CpG ODN treatment may influence the developmental life cycle of the parasites.
Collapse
Affiliation(s)
- Tajie H Harris
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin 53706, USA
| | | | | |
Collapse
|
28
|
Gazzinelli RT, Denkers EY. Protozoan encounters with Toll-like receptor signalling pathways: implications for host parasitism. Nat Rev Immunol 2006; 6:895-906. [PMID: 17110955 DOI: 10.1038/nri1978] [Citation(s) in RCA: 234] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Toll-like receptors (TLRs) have emerged as a major receptor family involved in non-self recognition. They have a vital role in triggering innate immunity and orchestrate the acquired immune response during bacterial and viral infection. However, the role of TLRs during infection with protozoan pathogens is less clear. Nevertheless, our understanding of how these parasitic microorganisms engage the host TLR signalling system has now entered a phase of rapid expansion. This Review describes recent insights into how parasitic protozoans are sensed by TLR molecules, and how the TLR system itself can be targeted by these microbial pathogens for their own survival.
Collapse
Affiliation(s)
- Ricardo T Gazzinelli
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, 31270-901 Belo Horizonte, Brazil.
| | | |
Collapse
|