101
|
Faezi F, Nahrevania H, Farahmand M, Sayyah M, Bidoki SK, Nemati S. Partial Immunotherapy of Leishmaniasis by in vivo Trial of L-Arginine in Balb/c Mice Infected with Leishmania major via Nitric Oxide Pathway. ACTA ACUST UNITED AC 2015. [DOI: 10.3923/ijbc.2015.110.122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
102
|
Lestinova T, Vlkova M, Votypka J, Volf P, Rohousova I. Phlebotomus papatasi exposure cross-protects mice against Leishmania major co-inoculated with Phlebotomus duboscqi salivary gland homogenate. Acta Trop 2015; 144:9-18. [PMID: 25597641 DOI: 10.1016/j.actatropica.2015.01.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 11/19/2014] [Accepted: 01/07/2015] [Indexed: 01/31/2023]
Abstract
Leishmania parasites are inoculated into host skin together with sand fly saliva and multiple exposures to uninfected sand fly bites protect mice against Leishmania infection. However, sand fly vectors differ in composition of the saliva and therefore the protection elicited by their salivary proteins was shown to be species-specific. On the other hand, the optimal vaccine based on sand fly salivary proteins should be based on conserved salivary proteins conferring cross-reactivity. In the present study we therefore focused on cross-protective properties of saliva from Phlebotomus papatasi and Phlebotomus duboscqi, the two natural vectors of Leishmania major. Two groups of mice exposed to bites of P. papatasi and two control, non-immunized groups were infected with L. major promastigotes along with either P. papatasi or P. duboscqi salivary gland homogenate. All mice were followed for the development of Leishmania lesions, parasite burdens, specific antibodies, and for production of NO, urea, or cytokines by peritoneal macrophages. Protection against Leishmania infection was observed not only in exposed mice challenged with homologous saliva but also in the group challenged with P. duboscqi saliva. Comparing both exposed groups, no significant differences were observed in parasite load, macrophage activity, or in the levels of anti-L. major and anti-P. papatasi/P. duboscqi antibodies. This is the first study showing cross-protection caused by salivary antigens of two Phlebotomus species. The cross-protective effect suggests that the anti-Leishmania vaccine based on P. papatasi salivary proteins might be applicable also in areas where L. major is transmitted by P. duboscqi.
Collapse
|
103
|
Muraille E, Leo O, Moser M. TH1/TH2 paradigm extended: macrophage polarization as an unappreciated pathogen-driven escape mechanism? Front Immunol 2014; 5:603. [PMID: 25505468 PMCID: PMC4244692 DOI: 10.3389/fimmu.2014.00603] [Citation(s) in RCA: 205] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 11/10/2014] [Indexed: 02/04/2023] Open
Abstract
The classical view of the Th1/Th2 paradigm posits that the pathogen nature, infectious cycle, and persistence represent key parameters controlling the choice of effector mechanisms operating during an immune response. Thus, efficient Th1 responses are triggered by replicating intracellular pathogens, while Th2 responses would control helminth infection and promote tissue repair during the resolution phase of an infectious event. However, this vision does not account for a growing body of data describing how pathogens exploit the polarization of the host immune response to their own benefit. Recently, the study of macrophages has illustrated a novel aspect of this arm race between pathogens and the immune system, and the central role of macrophages in homeostasis, repair and defense of all tissues is now fully appreciated. Like T lymphocytes, macrophages differentiate into distinct effectors including classically (M1) and alternatively (M2) activated macrophages. Interestingly, in addition to represent immune effectors, M1/M2 cells have been shown to represent potential reservoir cells to a wide range of intracellular pathogens. Subversion of macrophage cell metabolism by microbes appears as a recently uncovered immune escape strategy. Upon infection, several microbial agents have been shown to activate host metabolic pathways leading to the production of nutrients necessary to their long-term persistence in host. The purpose of this review is to summarize and discuss the strategies employed by pathogens to manipulate macrophage differentiation, and in particular their basic cell metabolism, to favor their own growth while avoiding immune control.
Collapse
Affiliation(s)
- Eric Muraille
- Laboratory of Parasitology, Faculty of Medicine, Université Libre de Bruxelles , Brussels , Belgium
| | - Oberdan Leo
- Laboratory of Immunobiology, Faculty of Sciences, Université Libre de Bruxelles , Gosselies , Belgium
| | - Muriel Moser
- Laboratory of Immunobiology, Faculty of Sciences, Université Libre de Bruxelles , Gosselies , Belgium
| |
Collapse
|
104
|
Roy S, Mukhopadhyay D, Mukherjee S, Ghosh S, Kumar S, Sarkar K, Pal D, Bhowmik P, Mandal K, Modak D, Guha SK, Pramanik N, Goswami RP, Saha B, Chatterjee M. A Defective Oxidative Burst and Impaired Antigen Presentation are Hallmarks of Human Visceral Leishmaniasis. J Clin Immunol 2014; 35:56-67. [PMID: 25479930 DOI: 10.1007/s10875-014-0115-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/30/2014] [Indexed: 01/18/2023]
Abstract
PURPOSE Survival of the Leishmania parasite within monocytes hinges on its ability to effectively nullify their microbicidal effector mechanisms. Accordingly, this study aimed to delineate this biological niche in patients with visceral leishmaniasis (VL). METHODS In monocytes, the redox status, antigen presenting capacity, expression of Toll-like receptors (TLRs), co-stimulatory molecules (CD80/86) and generation of intracellular cytokines (IL-8, IL-1β, IL-10 and LAP-TGF-β1) was measured by flow cytometry, levels of circulating cytokines (IL-1β, IL-6, TNF-α, IL-8, IL-4, IL-13, IL-10 and GM-CSF) by ELISA and arginase activity by spectrophotometry. RESULTS Within monocytes, generation of an oxidative burst was markedly attenuated as evident by decreased generation of nitric oxide and reactive oxygen species, concomitant with raised levels of thiols. This was accompanied by lowered frequency of TLR4(+) monocytes, but the arginase activity remained unaltered. Pathogen persistence was enhanced by the predominance of anti-inflammatory cytokines within monocytes, notably IL-10. Alongside, development of adaptive immunity was severely attenuated as manifested by a pronounced impairment of antigen presentation and co-stimulation evident by down regulation of CD54, HLA-DR and CD86. Treatment corrected the redox imbalance and reversed the impaired antigen presentation. CONCLUSIONS In VL, monocyte functions were severely impaired facilitating parasite persistence; anti-leishmanial chemotherapy mediated parasite elimination through modulation of the macrophage microenvironment by restoring its redox status and antigen presenting capacity.
Collapse
Affiliation(s)
- Susmita Roy
- Department of Pharmacology, Institute of Post Graduate Medical Education and Research, 244 B, Acharya J C Bose Road, Kolkata, 700020, West Bengal, India
| | - Debanjan Mukhopadhyay
- Department of Pharmacology, Institute of Post Graduate Medical Education and Research, 244 B, Acharya J C Bose Road, Kolkata, 700020, West Bengal, India
| | - Shibabrata Mukherjee
- Department of Pharmacology, Institute of Post Graduate Medical Education and Research, 244 B, Acharya J C Bose Road, Kolkata, 700020, West Bengal, India
| | - Susmita Ghosh
- Department of Pharmacology, Institute of Post Graduate Medical Education and Research, 244 B, Acharya J C Bose Road, Kolkata, 700020, West Bengal, India
| | - Shishir Kumar
- Department of Tropical Medicine, School of Tropical Medicine, Kolkata, 700073, West Bengal, India
| | - Kumkum Sarkar
- Department of Tropical Medicine, School of Tropical Medicine, Kolkata, 700073, West Bengal, India
| | - Dipankar Pal
- Department of Tropical Medicine, School of Tropical Medicine, Kolkata, 700073, West Bengal, India
| | - Pratik Bhowmik
- Department of Tropical Medicine, School of Tropical Medicine, Kolkata, 700073, West Bengal, India
| | - Kausik Mandal
- Department of Tropical Medicine, School of Tropical Medicine, Kolkata, 700073, West Bengal, India
| | - Dolanchampa Modak
- Department of Tropical Medicine, School of Tropical Medicine, Kolkata, 700073, West Bengal, India
| | - Subhasish Kamal Guha
- Department of Tropical Medicine, School of Tropical Medicine, Kolkata, 700073, West Bengal, India
| | - Netai Pramanik
- Department of Tropical Medicine, School of Tropical Medicine, Kolkata, 700073, West Bengal, India
| | - Rama Prosad Goswami
- Department of Tropical Medicine, School of Tropical Medicine, Kolkata, 700073, West Bengal, India
| | - Bibhuti Saha
- Department of Tropical Medicine, School of Tropical Medicine, Kolkata, 700073, West Bengal, India
| | - Mitali Chatterjee
- Department of Pharmacology, Institute of Post Graduate Medical Education and Research, 244 B, Acharya J C Bose Road, Kolkata, 700020, West Bengal, India.
| |
Collapse
|
105
|
Lamour SD, Veselkov KA, Posma JM, Giraud E, Rogers ME, Croft S, Marchesi JR, Holmes E, Seifert K, Saric J. Metabolic, Immune, and Gut Microbial Signals Mount a Systems Response to Leishmania major Infection. J Proteome Res 2014; 14:318-29. [DOI: 10.1021/pr5008202] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Sabrina D. Lamour
- Division
of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, Exhibition Road, London SW7 2AZ, United Kingdom
| | - Kirill A. Veselkov
- Division
of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, Exhibition Road, London SW7 2AZ, United Kingdom
| | - Joram M. Posma
- Division
of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, Exhibition Road, London SW7 2AZ, United Kingdom
| | - Emilie Giraud
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, United Kingdom
| | - Matthew E. Rogers
- Department of Disease Control, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, United Kingdom
| | - Simon Croft
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, United Kingdom
| | - Julian R. Marchesi
- Cardiff
School of Biosciences, Division of Microbiology, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, United Kingdom
- Centre
for Digestive and Gut Health, Imperial College London, Exhibition Road, London, SW7 2AZ, United Kingdom
| | - Elaine Holmes
- Division
of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, Exhibition Road, London SW7 2AZ, United Kingdom
| | - Karin Seifert
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, United Kingdom
| | - Jasmina Saric
- Division
of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, Exhibition Road, London SW7 2AZ, United Kingdom
| |
Collapse
|
106
|
Domínguez-Bernal G, Jiménez M, Molina R, Ordóñez-Gutiérrez L, Martínez-Rodrigo A, Mas A, Cutuli MT, Carrión J. Characterisation of the ex vivo virulence of Leishmania infantum isolates from Phlebotomus perniciosus from an outbreak of human leishmaniosis in Madrid, Spain. Parasit Vectors 2014; 7:499. [PMID: 25376381 PMCID: PMC4229600 DOI: 10.1186/s13071-014-0499-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 10/23/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Since mid 2009, an outbreak of human leishmaniosis in Madrid, Spain, has involved more than 560 clinical cases. Many of the cases occurred in people who live in areas around a newly constructed green park (BosqueSur). This periurban park provides a suitable habitat for sand flies (the vectors of Leishmania infantum). Indeed, studies of blood meals from sand flies captured in the area showed a strong association between the insect vector, hares or rabbits, and humans in the area. Interestingly, up to 70% of cases have been found in immunocompetent patients (aged between 46-60 years). This study was designed to evaluate the ex vivo virulence of the L. infantum isolates from Phlebotomus perniciosus captured in this area of Madrid. METHODS Murine macrophages and dendritic cells were infected ex vivo with L. infantum strain BCN150, isolate BOS1FL1, or isolate POL2FL7. At different times after infection, the infection indices, cytokine production (IL-12p40 and IL-10), NO release and arginase activities were evaluated. RESULTS Using an ex vivo model of infection in murine bone marrow-derived cells, we found that infection with isolates BOS1FL1 and POL2FL7 undermined host immune defence mechanisms in multiple ways. The main factors identified were changes in both the balance of iNOS versus arginase activities and the equilibrium between the production of IL-12 and IL-10. Infection with isolates BOS1FL1 and POL2FL7 also resulted in higher infection rates compared to the BCN150 strain. Infection index values at 24 h were as follows: BCN150-infected cells, 110 for infected MØ and 115 for infected DC; BOS1FL1-infected cells, 300 for infected MØ and 247 for infected DC; and POL2FL7-infected cells, 275 for infected MØ and 292 for infected DC. CONCLUSIONS Our data indicate that L. infantum isolates captured from this endemic area exhibited high virulence in terms of infection index, cytokine production and enzymatic activities involved in the pathogenesis of visceral leishmaniosis. Altogether, these data provide a starting point for the study of the virulence behaviour of parasites (BOS1FL1 and POL2FL7) isolated from P. perniciosus during the outbreak of human leishmaniosis in Madrid, Spain, and their involvement in infecting immunocompetent hosts.
Collapse
Affiliation(s)
- Gustavo Domínguez-Bernal
- Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040, Madrid, Spain.
| | - Maribel Jiménez
- Medical Entomology Unit, Department of Parasitology, National Centre of Microbiology, Carlos III Institute of Health, Majadahonda, 28220, Madrid, Spain.
| | - Ricardo Molina
- Medical Entomology Unit, Department of Parasitology, National Centre of Microbiology, Carlos III Institute of Health, Majadahonda, 28220, Madrid, Spain.
| | | | - Abel Martínez-Rodrigo
- Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040, Madrid, Spain.
| | - Alicia Mas
- Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040, Madrid, Spain.
| | - Maria Teresa Cutuli
- Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040, Madrid, Spain.
| | - Javier Carrión
- Department of Animal Health, Faculty of Veterinary Science, Complutense University of Madrid, 28040, Madrid, Spain.
| |
Collapse
|
107
|
Rath M, Müller I, Kropf P, Closs EI, Munder M. Metabolism via Arginase or Nitric Oxide Synthase: Two Competing Arginine Pathways in Macrophages. Front Immunol 2014; 5:532. [PMID: 25386178 PMCID: PMC4209874 DOI: 10.3389/fimmu.2014.00532] [Citation(s) in RCA: 784] [Impact Index Per Article: 78.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 10/09/2014] [Indexed: 12/23/2022] Open
Abstract
Macrophages play a major role in the immune system, both as antimicrobial effector cells and as immunoregulatory cells, which induce, suppress or modulate adaptive immune responses. These key aspects of macrophage biology are fundamentally driven by the phenotype of macrophage arginine metabolism that is prevalent in an evolving or ongoing immune response. M1 macrophages express the enzyme nitric oxide synthase, which metabolizes arginine to nitric oxide (NO) and citrulline. NO can be metabolized to further downstream reactive nitrogen species, while citrulline might be reused for efficient NO synthesis via the citrulline–NO cycle. M2 macrophages are characterized by expression of the enzyme arginase, which hydrolyzes arginine to ornithine and urea. The arginase pathway limits arginine availability for NO synthesis and ornithine itself can further feed into the important downstream pathways of polyamine and proline syntheses, which are important for cellular proliferation and tissue repair. M1 versus M2 polarization leads to opposing outcomes of inflammatory reactions, but depending on the context, M1 and M2 macrophages can be both pro- and anti-inflammatory. Notably, M1/M2 macrophage polarization can be driven by microbial infection or innate danger signals without any influence of adaptive immune cells, secondarily driving the T helper (Th)1/Th2 polarization of the evolving adaptive immune response. Since both arginine metabolic pathways cross-inhibit each other on the level of the respective arginine break-down products and Th1 and Th2 lymphocytes can drive or amplify macrophage M1/M2 dichotomy via cytokine activation, this forms the basis of a self-sustaining M1/M2 polarization of the whole immune response. Understanding the arginine metabolism of M1/M2 macrophage phenotypes is therefore central to find new possibilities to manipulate immune responses in infection, autoimmune diseases, chronic inflammatory conditions, and cancer.
Collapse
Affiliation(s)
- Meera Rath
- Department of Pharmacology, Institute of Medical Sciences, Faculty of Medical Sciences, Siksha 'O' Anusandhan University , Bhubaneshwar , India
| | - Ingrid Müller
- Section of Immunology, Department of Medicine, Imperial College London , London , UK
| | - Pascale Kropf
- Section of Immunology, Department of Medicine, Imperial College London , London , UK
| | - Ellen I Closs
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University , Mainz , Germany
| | - Markus Munder
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center, Johannes Gutenberg University , Mainz , Germany ; Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University , Mainz , Germany
| |
Collapse
|
108
|
Yang Z, Ming XF. Functions of arginase isoforms in macrophage inflammatory responses: impact on cardiovascular diseases and metabolic disorders. Front Immunol 2014; 5:533. [PMID: 25386179 PMCID: PMC4209887 DOI: 10.3389/fimmu.2014.00533] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 10/09/2014] [Indexed: 12/18/2022] Open
Abstract
Macrophages play a paramount role in immunity and inflammation-associated diseases, including infections, cardiovascular diseases, obesity-associated metabolic imbalances, and cancer. Compelling evidence from studies of recent years demonstrates that macrophages are heterogeneous and undergo heterogeneous phenotypic changes in response to microenvironmental stimuli. The M1 killer type response and the M2 repair type response are best known, and are two extreme examples. Among other markers, inducible nitric oxide synthase and type-I arginase (Arg-I), the enzymes that are involved in l-arginine/nitric oxide (NO) metabolism, are associated with the M1 and M2 phenotype, respectively, and therefore widely used as the markers for characterization of the two macrophage phenotypes. There is also a type-II arginase (Arg-II), which is expressed in macrophages and prevalently viewed as having the same function as Arg-I in the cells. In contrast to Arg-I, little information on the role of Arg-II in macrophage inflammatory responses is available. Emerging evidence, however, suggests that differential roles of Arg-I and Arg-II in regulating macrophage functions. In this article, we will review recent developments on the functional roles of the two arginase isoforms in regulation of macrophage inflammatory responses by focusing on their impact on the pathogenesis of cardiovascular diseases and metabolic disorders.
Collapse
Affiliation(s)
- Zhihong Yang
- Vascular Biology, Division of Physiology, Department of Medicine, Faculty of Science, University of Fribourg , Fribourg , Switzerland
| | - Xiu-Fen Ming
- Vascular Biology, Division of Physiology, Department of Medicine, Faculty of Science, University of Fribourg , Fribourg , Switzerland
| |
Collapse
|
109
|
Corware K, Yardley V, Mack C, Schuster S, Al-Hassi H, Herath S, Bergin P, Modolell M, Munder M, Müller I, Kropf P. Protein energy malnutrition increases arginase activity in monocytes and macrophages. Nutr Metab (Lond) 2014; 11:51. [PMID: 25392710 PMCID: PMC4228191 DOI: 10.1186/1743-7075-11-51] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 09/24/2014] [Indexed: 11/24/2022] Open
Abstract
Background Protein energy malnutrition is commonly associated with immune dysfunctions and is a major factor in susceptibility to infectious diseases. Methods In this study, we evaluated the impact of protein energy malnutrition on the capacity of monocytes and macrophages to upregulate arginase, an enzyme associated with immunosuppression and increased pathogen replication. Results Our results show that monocytes and macrophages are significantly increased in the bone marrow and blood of mice fed on a protein low diet. No alteration in the capacity of bone marrow derived macrophages isolated from malnourished mice to phagocytose particles, to produce the microbicidal molecule nitric oxide and to kill intracellular Leishmania parasites was detected. However, macrophages and monocytes from malnourished mice express significantly more arginase both in vitro and in vivo. Using an experimental model of visceral leishmaniasis, we show that following protein energy malnutrition, the increased parasite burden measured in the spleen of these mice coincided with increased arginase activity and that macrophages provide a more permissive environment for parasite growth. Conclusions Taken together, these results identify a novel mechanism in protein energy malnutrition that might contributes to increased susceptibility to infectious diseases by upregulating arginase activity in myeloid cells. Electronic supplementary material The online version of this article (doi:10.1186/1743-7075-11-51) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Karina Corware
- Department of Medicine, Section of Immunology, Faculty of Medicine, Imperial College London, Norfolk Place, London, W2 1PG UK
| | - Vanessa Yardley
- Immunology and Infection Department, London School of Hygiene and Tropical Medicine, London, UK
| | - Christopher Mack
- Department of Medicine, Section of Immunology, Faculty of Medicine, Imperial College London, Norfolk Place, London, W2 1PG UK
| | - Steffen Schuster
- Department of Biochemistry, WHO Immunology Research and Training Center, University of Lausanne, Lausanne, Switzerland
| | - Hafid Al-Hassi
- Department of Medicine, Section of Immunology, Faculty of Medicine, Imperial College London, Norfolk Place, London, W2 1PG UK
| | - Shanthi Herath
- School of Biological Sciences, Royal Holloway, University of London, Egham, UK
| | - Philip Bergin
- International AIDS Vaccine Initiative Human Immunology Laboratory, Faculty of Medicine, Imperial College London, London, UK
| | - Manuel Modolell
- Department of Cellular Immunology, Max-Planck-Institute for Immunobiology and Epigenetics, Freiburg, Germany
| | - Markus Munder
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center Mainz, Mainz, Germany
| | - Ingrid Müller
- Department of Medicine, Section of Immunology, Faculty of Medicine, Imperial College London, Norfolk Place, London, W2 1PG UK
| | - Pascale Kropf
- Department of Medicine, Section of Immunology, Faculty of Medicine, Imperial College London, Norfolk Place, London, W2 1PG UK
| |
Collapse
|
110
|
Macrophage arginase-1 controls bacterial growth and pathology in hypoxic tuberculosis granulomas. Proc Natl Acad Sci U S A 2014; 111:E4024-32. [PMID: 25201986 DOI: 10.1073/pnas.1408839111] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Lung granulomas develop upon Mycobacterium tuberculosis (Mtb) infection as a hallmark of human tuberculosis (TB). They are structured aggregates consisting mainly of Mtb-infected and -uninfected macrophages and Mtb-specific T cells. The production of NO by granuloma macrophages expressing nitric oxide synthase-2 (NOS2) via l-arginine and oxygen is a key protective mechanism against mycobacteria. Despite this protection, TB granulomas are often hypoxic, and bacterial killing via NOS2 in these conditions is likely suboptimal. Arginase-1 (Arg1) also metabolizes l-arginine but does not require oxygen as a substrate and has been shown to regulate NOS2 via substrate competition. However, in other infectious diseases in which granulomas occur, such as leishmaniasis and schistosomiasis, Arg1 plays additional roles such as T-cell regulation and tissue repair that are independent of NOS2 suppression. To address whether Arg1 could perform similar functions in hypoxic regions of TB granulomas, we used a TB murine granuloma model in which NOS2 is absent. Abrogation of Arg1 expression in macrophages in this setting resulted in exacerbated lung granuloma pathology and bacterial burden. Arg1 expression in hypoxic granuloma regions correlated with decreased T-cell proliferation, suggesting that Arg1 regulation of T-cell immunity is involved in disease control. Our data argue that Arg1 plays a central role in the control of TB when NOS2 is rendered ineffective by hypoxia.
Collapse
|
111
|
Abu-Lubad M, Meyer TF, Al-Zeer MA. Chlamydia trachomatis inhibits inducible NO synthase in human mesenchymal stem cells by stimulating polyamine synthesis. THE JOURNAL OF IMMUNOLOGY 2014; 193:2941-51. [PMID: 25114102 DOI: 10.4049/jimmunol.1400377] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Chlamydia trachomatis is considered the most common agent of sexually transmitted disease worldwide. As an obligate intracellular bacterium, it relies on the host for survival. Production of NO is an effective antimicrobial defense mechanism of the innate immune system. However, whether NO is able to arrest chlamydial growth remains unclear. Similarly, little is known about the mechanisms underlying subversion of cellular innate immunity by C. trachomatis. By analyzing protein and mRNA expression in infected human mesenchymal stem cells, combined with RNA interference and biochemical assays, we observed that infection with C. trachomatis led to downregulated expression of inducible NO synthase (iNOS) in human mesenchymal stem cells in vitro. Furthermore, infection upregulated the expression of the rate-limiting enzyme in the polyamine biosynthetic pathway, ornithine decarboxylase, diverting the iNOS substrate l-arginine toward the synthesis of polyamines. Inhibition of ornithine decarboxylase activity using small interfering RNA or the competitive inhibitor difluoromethylornithine restored iNOS protein expression and activity in infected cells and inhibited chlamydial growth. This inhibition was mediated through tyrosine nitration of chlamydial protein by peroxynitrite, an NO metabolite. Thus, Chlamydia evades innate immunity by inhibiting NO production through induction of the alternative polyamine pathway.
Collapse
Affiliation(s)
- Mohammad Abu-Lubad
- Department of Molecular Biology, Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| | - Thomas F Meyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| | - Munir A Al-Zeer
- Department of Molecular Biology, Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| |
Collapse
|
112
|
Resveratrol is active against Leishmania amazonensis: in vitro effect of its association with Amphotericin B. Antimicrob Agents Chemother 2014; 58:6197-208. [PMID: 25114129 DOI: 10.1128/aac.00093-14] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Resveratrol is a polyphenol found in black grapes and red wine and has many biological activities. In this study, we evaluated the effect of resveratrol alone and in association with amphotericin B (AMB) against Leishmania amazonensis. Our results demonstrate that resveratrol possesses both antipromastigote and antiamastigote effects, with 50% inhibitory concentrations (IC50s) of 27 and 42 μM, respectively. The association of resveratrol with AMB showed synergy for L. amazonensis amastigotes, as demonstrated by the mean sums of fractional inhibitory index concentration (mean ΣFIC) of 0.483, although for promastigotes, this association was indifferent. Treatment with resveratrol increased the percentage of promastigotes in the sub-G0/G1 phase of the cell cycle, reduced the mitochondrial potential, and showed an elevated choline peak and CH2-to-CH3 ratio in the nuclear magnetic resonance (NMR) spectroscopy analysis; all these features indicate parasite death. Resveratrol also decreased the activity of the enzyme arginase in uninfected and infected macrophages with and without stimulation with interleukin-4 (IL-4), also implicating arginase inhibition in parasite death. The anti-Leishmania effect of resveratrol and its potential synergistic association with AMB indicate that these compounds should be subjected to further studies of drug association therapy in vivo.
Collapse
|
113
|
Heitmann L, Abad Dar M, Schreiber T, Erdmann H, Behrends J, Mckenzie ANJ, Brombacher F, Ehlers S, Hölscher C. The IL-13/IL-4Rα axis is involved in tuberculosis-associated pathology. J Pathol 2014; 234:338-50. [PMID: 24979482 PMCID: PMC4277691 DOI: 10.1002/path.4399] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 06/05/2014] [Accepted: 06/22/2014] [Indexed: 01/20/2023]
Abstract
Human tuberculosis (TB) is a leading global health threat and still constitutes a major medical challenge. However, mechanisms governing tissue pathology during post-primary TB remain elusive, partly because genetically or immunologically tractable animal models are lacking. In human TB, the demonstration of a large relative increase in interleukin (IL)-4 and IL-13 expression, which correlates with lung damage, indicates that a subversive T helper (TH)2 component in the response to Mycobacterium tuberculosis (Mtb) may undermine protective immunity and contribute to reactivation and tissue pathology. Up to now, there has been no clear evidence regarding whether IL-4/IL-13-IL-4 receptor-α (Rα)-mediated mechanisms may in fact cause reactivation and pathology. Unfortunately, the virtual absence of centrally necrotizing granulomas in experimental murine TB is associated with a poor induction of a TH2 immune response. We therefore hypothesize that, in mice, an increased production of IL-13 may lead to a pathology similar to human post-primary TB. In our study, aerosol Mtb infection of IL-13-over-expressing mice in fact resulted in pulmonary centrally necrotizing granulomas with multinucleated giant cells, a hypoxic rim and a perinecrotic collagen capsule, with an adjacent zone of lipid-rich, acid-fast bacilli-containing foamy macrophages, thus strongly resembling the pathology in human post-primary TB. Granuloma necrosis (GN) in Mtb-infected IL-13-over-expressing mice was associated with the induction of arginase-1-expressing macrophages. Indirect blockade of the endogenous arginase inhibitor l-hydroxyarginine in Mtb-infected wild-type mice resulted in a strong arginase expression and precipitated a similar pathology of GN. Together, we here introduce an experimental TB model that displays many features of centrally necrotizing granulomas in human post-primary TB and demonstrate that IL-13/IL-4Rα-dependent mechanisms leading to arginase-1 expression are involved in TB-associated tissue pathology.
Collapse
Affiliation(s)
- Lisa Heitmann
- Infection Immunology, Research Centre Borstel, Germany; Priority Research Area 'Infections', Research Centre Borstel, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Osorio EY, Travi BL, da Cruz AM, Saldarriaga OA, Medina AA, Melby PC. Growth factor and Th2 cytokine signaling pathways converge at STAT6 to promote arginase expression in progressive experimental visceral leishmaniasis. PLoS Pathog 2014; 10:e1004165. [PMID: 24967908 PMCID: PMC4072777 DOI: 10.1371/journal.ppat.1004165] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 04/21/2014] [Indexed: 12/26/2022] Open
Abstract
Host arginase 1 (arg1) expression is a significant contributor to the pathogenesis of progressive visceral leishmaniasis (VL), a neglected tropical disease caused by the intracellular protozoan Leishmania donovani. Previously we found that parasite-induced arg1 expression in macrophages was dependent on STAT6 activation. Arg1 expression was amplified by, but did not require, IL-4, and required de novo synthesis of unknown protein(s). To further explore the mechanisms involved in arg1 regulation in VL, we screened a panel of kinase inhibitors and found that inhibitors of growth factor signaling reduced arg1 expression in splenic macrophages from hamsters with VL. Analysis of growth factors and their signaling pathways revealed that the Fibroblast Growth Factor Receptor 1 (FGFR-1) and Insulin-like Growth Factor 1 Receptor (IGF-1R) and a number of downstream signaling proteins were activated in splenic macrophages isolated from hamsters infected with L. donovani. Recombinant FGF-2 and IGF-1 increased the expression of arg1 in L. donovani infected hamster macrophages, and this induction was augmented by IL-4. Inhibition of FGFR-1 and IGF-1R decreased arg1 expression and restricted L. donovani replication in both in vitro and ex vivo models of infection. Inhibition of the downstream signaling molecules JAK and AKT also reduced the expression of arg1 in infected macrophages. STAT6 was activated in infected macrophages exposed to either FGF-2 or IGF-1, and STAT6 was critical to the FGFR-1- and IGF-1R-mediated expression of arg1. The converse was also true as inhibition of FGFR-1 and IGF-1R reduced the activation of STAT6 in infected macrophages. Collectively, these data indicate that the FGFR/IGF-1R and IL-4 signaling pathways converge at STAT6 to promote pathologic arg1 expression and intracellular parasite survival in VL. Targeted interruption of these pathological processes offers an approach to restrain this relentlessly progressive disease. Visceral leishmaniasis (VL), caused by the intracellular protozoan Leishmania donovani, is a progressive infection that is particularly common in impoverished populations of the world. People die from this disease unless it is treated. We used an experimental infection model that mimics the clinical and pathological features of human VL to study how the parasite causes this severe disease. We found that host macrophages infected with Leishmania donovani are activated in a way that leads to the expression of arginase, an enzyme that counteracts the cell's mechanisms that control the infection. This disease-promoting activation pathway was driven by the convergence of growth factor and cytokine signaling pathways and activation of the transcription factor STAT6. Chemical inhibition of signaling through the fibroblast growth factor receptor-1 (FGFR-1) or insulin-like growth factor-1 receptor (IGF-IR), or genetic knockdown of STAT6 led to reduced expression of arginase and enhanced control of the infection by macrophages. This indicates that the growth factor signaling pathways together with the cytokine pathways promote this disease. Interventions designed to disrupt this signaling could help in the treatment of VL.
Collapse
Affiliation(s)
- E Yaneth Osorio
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America; Laboratório Interdisciplinar de Pesquisas Médicas (LIPMED), Instituto Oswaldo Cruz-FIOCRUZ, Rio de Janeiro, Brazil
| | - Bruno L Travi
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Alda M da Cruz
- Laboratório Interdisciplinar de Pesquisas Médicas (LIPMED), Instituto Oswaldo Cruz-FIOCRUZ, Rio de Janeiro, Brazil
| | - Omar A Saldarriaga
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Audrie A Medina
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Peter C Melby
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America; Center for Tropical Diseases, and Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America; Department of Pathology, and Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
115
|
Sousa LMA, Carneiro MBH, Resende ME, Martins LS, Dos Santos LM, Vaz LG, Mello PS, Mosser DM, Oliveira MAP, Vieira LQ. Neutrophils have a protective role during early stages of Leishmania amazonensis infection in BALB/c mice. Parasite Immunol 2014; 36:13-31. [PMID: 24102495 DOI: 10.1111/pim.12078] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 08/05/2013] [Indexed: 01/13/2023]
Abstract
Neutrophils are involved in the early stages of immune responses to pathogens. Here, we investigated the role of neutrophils during the establishment of Leishmania amazonensis infection in BALB/c and C57BL/6 mice. First, we showed an accumulation of neutrophils between 6 and 24 h post-infection, followed by a reduction in neutrophil numbers after 72 h. Next, we depleted neutrophils prior to infection using RB6-8C5 or 1A8 mAb. Neutrophil depletion led to faster lesion development, increased parasite numbers and higher arginase activity during the first week of infection in BALB/c mice, but not in C57BL/6 mice. Increased susceptibility was accompanied by augmented levels of anti-L. amazonensis IgG and increased production of IL-10 and IL-17. Because IL-10 is a mediator of susceptibility to Leishmania infection, we blocked IL-10 signalling in neutrophil-depleted mice using anti-IL-10R. Interestingly, inhibition of IL-10 signalling abrogated the increase in parasite loads observed in neutrophil-depleted mice, suggesting that parasite proliferation is at least partially mediated by IL-10. Additionally, we tested the effect of IL-17 in inflammatory macrophages and observed that IL-17 increased arginase activity and favoured parasite growth. Taken together, our data indicate that neutrophils control parasite numbers and limit lesion development during the first week of infection in BALB/c mice.
Collapse
Affiliation(s)
- L M A Sousa
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Jang JC, Nair MG. Alternatively Activated Macrophages Revisited: New Insights into the Regulation of Immunity, Inflammation and Metabolic Function following Parasite Infection. ACTA ACUST UNITED AC 2014; 9:147-156. [PMID: 24772059 DOI: 10.2174/1573395509666131210232548] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The role of macrophages in homeostatic conditions and the immune system range from clearing debris to recognizing and killing pathogens. While classically activated macrophages (CAMacs) are induced by T helper type 1 (Th1) cytokines and exhibit microbicidal properties, Th2 cytokines promote alternative activation of macrophages (AAMacs). AAMacs contribute to the killing of helminth parasites and mediate additional host-protective processes such as regulating inflammation and wound healing. Yet, other parasites susceptible to Th1 type responses can exploit alternative activation of macrophages to diminish Th1 immune responses and prolong infection. In this review, we will delineate the factors that mediate alternative activation (e.g. Th2 cytokines and chitin) and the resulting downstream signaling events (e.g. STAT6 signaling). Next, the specific AAMac-derived factors (e.g. Arginase1) that contribute to resistance or susceptibility to parasitic infections will be summarized. Finally, we will conclude with the discussion of additional AAMac functions beyond immunity to parasites, including the regulation of inflammation, wound healing and the regulation of metabolic disorders.
Collapse
Affiliation(s)
- Jessica C Jang
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA
| | - Meera G Nair
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA
| |
Collapse
|
117
|
Abstract
The presence of different sets of several enzymes that participate in the Krebs-Henseleit cycle has been used to identify several genera of trypanosomatids. One of these enzymes is arginase (L-arginine amidinohydrolase, E.C. 3.5.3.1), a metalloenzyme that catalyzes the hydrolysis of L-arginine to L-ornithine and urea. Arginase activity has been detected in Leishmania, Crithidia and Leptomonas but not in Trypanosoma, Herpetomonas or Phytomonas. The ureotelic behavior of some trypanosomatids is not due to urea excretion but to the production of ornithine to supply the polyamine pathway, which is essential for replication. Leishmania is found inside macrophages in the mammalian host and to live in these cells, the parasite must escape from several microbicidal mechanisms, such as nitric oxide (NO) production mediated by inducible nitric oxide synthase (iNOS). Since arginase and iNOS use the L-arginine as substrate, the amount of this amino acid available for both pathways is critical for parasite replication. In both promastigotes and amastigotes, arginase is located in the glycosome indicating that arginine trafficking in the cell is used to provide the optimal concentration of substrate for arginase. Arginine uptake by the parasite is also important in supplying the arginase substrate. Leishmania responds to arginine starvation by increasing the amino acid uptake. In addition to the external supply, the internal L-arginine pool also governs the uptake of this amino acid, and the size of this internal pool is modulated by arginase activity. Thus, arginine uptake and arginase activity are important in establishing and maintaining Leishmania infection.
Collapse
|
118
|
Lisi L, Tramutola A, Navarra P, Dello Russo C. Antiretroviral agents increase NO production in gp120/IFNγ-stimulated cultures of rat microglia via an arginase-dependent mechanism. J Neuroimmunol 2014; 266:24-32. [DOI: 10.1016/j.jneuroim.2013.10.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 10/31/2013] [Accepted: 10/31/2013] [Indexed: 12/22/2022]
|
119
|
Abstract
In the healthy gastrointestinal tract, homeostasis is an active process that requires a careful balance of host responses to the enteric luminal contents. Intestinal macrophages and dendritic cells (DCs) comprise a unique group of tissue immune cells that are ideally situated at the interface of the host and the enteric luminal environment to appropriately respond to microbes and ingested stimuli. However, intrinsic defects in macrophage and DC function contribute to the pathogenesis of inflammatory bowel diseases, as highlighted by recent genome-wide association studies. Gastrointestinal macrophages and DCs participate in inflammatory bowel disease development through inappropriate responses to enteric microbial stimuli, inefficient clearance of microbes from host tissues, and impaired transition from appropriate proinflammatory responses to anti-inflammatory responses that promote resolution. By understanding how intestinal macrophages and DCs initiate chronic inflammation, new pathogenesis-based therapeutic strategies to treat human inflammatory bowel diseases will be elucidated.
Collapse
|
120
|
Deletion of IL-4 receptor alpha on dendritic cells renders BALB/c mice hypersusceptible to Leishmania major infection. PLoS Pathog 2013; 9:e1003699. [PMID: 24204259 PMCID: PMC3812013 DOI: 10.1371/journal.ppat.1003699] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 08/28/2013] [Indexed: 02/05/2023] Open
Abstract
In BALB/c mice, susceptibility to infection with the intracellular parasite Leishmania major is driven largely by the development of T helper 2 (Th2) responses and the production of interleukin (IL)-4 and IL-13, which share a common receptor subunit, the IL-4 receptor alpha chain (IL-4Rα). While IL-4 is the main inducer of Th2 responses, paradoxically, it has been shown that exogenously administered IL-4 can promote dendritic cell (DC) IL-12 production and enhance Th1 development if given early during infection. To further investigate the relevance of biological quantities of IL-4 acting on DCs during in vivo infection, DC specific IL-4Rα deficient (CD11c(cre)IL-4Rα(-/lox)) BALB/c mice were generated by gene targeting and site-specific recombination using the cre/loxP system under control of the cd11c locus. DNA, protein, and functional characterization showed abrogated IL-4Rα expression on dendritic cells and alveolar macrophages in CD11c(cre)IL-4Rα(-/lox) mice. Following infection with L. major, CD11c(cre)IL-4Rα(-/lox) mice became hypersusceptible to disease, presenting earlier and increased footpad swelling, necrosis and parasite burdens, upregulated Th2 cytokine responses and increased type 2 antibody production as well as impaired classical activation of macrophages. Hypersusceptibility in CD11c(cre)IL-4Rα(-/lox) mice was accompanied by a striking increase in parasite burdens in peripheral organs such as the spleen, liver, and even the brain. DCs showed increased parasite loads in CD11c(cre)IL-4Rα(-/lox) mice and reduced iNOS production. IL-4Rα-deficient DCs produced reduced IL-12 but increased IL-10 due to impaired DC instruction, with increased mRNA expression of IL-23p19 and activin A, cytokines previously implicated in promoting Th2 responses. Together, these data demonstrate that abrogation of IL-4Rα signaling on DCs is severely detrimental to the host, leading to rapid disease progression, and increased survival of parasites in infected DCs due to reduced killing effector functions.
Collapse
|
121
|
Moura VBL, Silva MM, Batista LF, Gomes CM, Leenen PJM, Lino RS, Oliveira MAP. Arginase activity is associated with fibrosis in experimental infection with Taenia crassiceps, but does not play a major role in resistance to infection. Exp Parasitol 2013; 135:599-605. [PMID: 24090570 DOI: 10.1016/j.exppara.2013.09.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 09/16/2013] [Accepted: 09/22/2013] [Indexed: 12/01/2022]
Abstract
Murine infection with Taenia crassiceps cysticerci is used as an experimental model for human and animal cysticercosis. In this infection parasites can be found associated with an inflammatory infiltrate enriched with macrophages. Experimental evidence exists supporting a role for either NO-producing classically activated (CAMΦ) or arginase- and CD301-expressing alternatively activated macrophages (AAMΦ) in T. crassiceps resistance. In both cell types, arginine is utilized as an important mediator in macrophage effector functions. To investigate whether there is an association between arginine availability, susceptibility to T. crassiceps and other parameters such as fibrosis, BALB/c mice were infected intraperitoneally with cysticerci and treated daily with the arginase inhibitor nor-NOHA or supplemented with l-arginine and followed for eight weeks. The numbers and developmental stages of parasites were evaluated as well as the presence of CD301+ AAMΦ, arginase activity and collagen deposition in the peritoneal membrane. Treatment with the arginase inhibitor or supplementation with l-arginine did not change the parasitic load or profile of the infection. However, the arginase inhibitor significantly decreased the deposition of collagen. These results suggest that arginase activity does not interfere with parasite control during experimental infection with T. crassiceps, but it is important for fibrosis in cysticercosis.
Collapse
Affiliation(s)
- Vania B L Moura
- Department of Microbiology, Immunology, Parasitology and Pathology, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Rua 235 S/N, Setor Universitário, 74605-050 Goiânia, Goiás, Brazil.
| | | | | | | | | | | | | |
Collapse
|
122
|
MAP kinase phosphatase-2 plays a key role in the control of infection with Toxoplasma gondii by modulating iNOS and arginase-1 activities in mice. PLoS Pathog 2013; 9:e1003535. [PMID: 23966857 PMCID: PMC3744406 DOI: 10.1371/journal.ppat.1003535] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 06/18/2013] [Indexed: 11/19/2022] Open
Abstract
The dual specific phosphatase, MAP kinase phosphatase-2 (MKP-2) has recently been demonstrated to negatively regulate macrophage arginase-1 expression, while at the same time to positively regulate iNOS expression. Consequently, MKP-2 is likely to play a significant role in the host interplay with intracellular pathogens. Here we demonstrate that MKP-2(-/-) mice on the C57BL/6 background have enhanced susceptibility compared with wild-type counterparts following infection with type-2 strains of Toxoplasma gondii as measured by increased parasite multiplication during acute infection, increased mortality from day 12 post-infection onwards and increased parasite burdens in the brain, day 30 post-infection. MKP-2(-/-) mice did not, however, demonstrate defective type-1 responses compared with MKP-2(+/+) mice following infection although they did display significantly reduced serum nitrite levels and enhanced tissue arginase-1 expression. Early resistance to T. gondii in MKP-2(+/+), but not MKP-2(-/-), mice was nitric oxide (NO) dependent as infected MKP-2(+/+), but not MKP-2(-/-) mice succumbed within 10 days post-infection with increased parasite burdens following treatment with the iNOS inhibitor L-NAME. Conversely, treatment of infected MKP-2(-/-) but not MKP-2(+/+) mice with nor-NOHA increased parasite burdens indicating a protective role for arginase-1 in MKP-2(-/-) mice. In vitro studies using tachyzoite-infected bone marrow derived macrophages and selective inhibition of arginase-1 and iNOS activities confirmed that both iNOS and arginase-1 contributed to inhibiting parasite replication. However, the effects of arginase-1 were transient and ultimately the role of iNOS was paramount in facilitating long-term inhibition of parasite multiplication within macrophages.
Collapse
|
123
|
D'Antonio EL, Ullman B, Roberts SC, Dixit UG, Wilson ME, Hai Y, Christianson DW. Crystal structure of arginase from Leishmania mexicana and implications for the inhibition of polyamine biosynthesis in parasitic infections. Arch Biochem Biophys 2013; 535:163-76. [PMID: 23583962 DOI: 10.1016/j.abb.2013.03.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 03/25/2013] [Accepted: 03/26/2013] [Indexed: 02/06/2023]
Abstract
Arginase from parasitic protozoa belonging to the genus Leishmania is a potential drug target for the treatment of leishmaniasis because this binuclear manganese metalloenzyme catalyzes the first committed step in the biosynthesis of polyamines that enable cell growth and survival. The high resolution X-ray crystal structures of the unliganded form of Leishmania mexicana arginase (LmARG) and four inhibitor complexes are now reported. These complexes include the reactive substrate analogue 2(S)-amino-6-boronohexanoic acid (ABH) and the hydroxylated substrate analogue nor-N(ω)-hydroxy-l-arginine (nor-NOHA), which are the most potent arginase inhibitors known to date. Comparisons of the LmARG structure with that of the archetypal arginase, human arginase I, reveal that all residues important for substrate binding and catalysis are strictly conserved. However, three regions of tertiary structure differ between the parasitic enzyme and the human enzyme corresponding to the G62 - S71, L161 - C172, and I219 - V230 segments of LmARG. Additionally, variations are observed in salt link interactions that stabilize trimer assembly in LmARG. We also report biological studies in which we demonstrate that localization of LmARG to the glycosome, a unique subcellular organelle peculiar to Leishmania and related parasites, is essential for robust pathogenesis.
Collapse
Affiliation(s)
- Edward L D'Antonio
- Roy and Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, PA 19104-6323, USA
| | | | | | | | | | | | | |
Collapse
|
124
|
Arginase inhibition prevents the low shear stress-induced development of vulnerable atherosclerotic plaques in ApoE−/− mice. Atherosclerosis 2013; 227:236-43. [DOI: 10.1016/j.atherosclerosis.2012.12.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 12/09/2012] [Accepted: 12/15/2012] [Indexed: 02/03/2023]
|
125
|
Kling JC, Körner H. Different regulatory mechanisms in protozoan parasitic infections. Int J Parasitol 2013; 43:417-25. [PMID: 23470812 DOI: 10.1016/j.ijpara.2013.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 02/01/2013] [Accepted: 02/04/2013] [Indexed: 02/07/2023]
Abstract
The immune response to the protozoan pathogens, Leishmania spp., Trypanosoma spp. and Plasmodium spp., has been studied extensively with particular focus on regulation of the immune response by immunological mechanisms. More specifically, in diseases caused by parasites, immunosuppression frequently prevents immunopathology that can injure the host. However, this allows a small number of parasites to evade the immune response and remain in the host after a clinical cure. The consequences can be chronic infections, which establish a zoonotic or anthroponotic reservoir. This review will highlight some of the identified regulatory mechanisms of the immune system that govern immune responses to parasitic diseases, in particular leishmaniasis, trypanosomiasis and malaria, and discuss implications for the development of efficient vaccines against these diseases.
Collapse
Affiliation(s)
- Jessica C Kling
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania 7000, Australia
| | | |
Collapse
|
126
|
Sans-Fons MG, Yeramian A, Pereira-Lopes S, Santamaría-Babi LF, Modolell M, Lloberas J, Celada A. Arginine transport is impaired in C57Bl/6 mouse macrophages as a result of a deletion in the promoter of Slc7a2 (CAT2), and susceptibility to Leishmania infection is reduced. J Infect Dis 2013; 207:1684-93. [PMID: 23460752 DOI: 10.1093/infdis/jit084] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Host genetic factors play a crucial role in immune response. To determine whether the differences between C57Bl/6 and BALB-C mice are due only to the production of cytokines by T-helper 1 cells or T-helper 2 cells, we obtained bone marrow-derived macrophages from both strains and incubated them with these cytokines. Although the induction of Nos2 and Arg1 was similar in the 2 strains, infectivity to Leishmania major differed, as did macrophage uptake of arginine, which was higher in BALB-C macrophages. The levels of interferon γ- and interleukin 4-dependent induction of the cationic amino acid transporter SLC7A2 (also known as "cationic amino acid transporter 2," or "CAT2") were decreased in macrophages from C57Bl/6 mice. This reduction was a result of a deletion in the promoter of one of the 4 AGGG repeats. These results demonstrate that the availability of arginine controls critical aspects of macrophage activation and reveal a factor for susceptibility to Leishmania infection.
Collapse
Affiliation(s)
- M Gloria Sans-Fons
- Macrophage Biology Group, Department of Physiology and Immunology, Universitat de Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
127
|
Mou Z, Muleme HM, Liu D, Jia P, Okwor IB, Kuriakose SM, Beverley SM, Uzonna JE. Parasite-derived arginase influences secondary anti-Leishmania immunity by regulating programmed cell death-1-mediated CD4+ T cell exhaustion. THE JOURNAL OF IMMUNOLOGY 2013; 190:3380-9. [PMID: 23460745 DOI: 10.4049/jimmunol.1202537] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The breakdown of L-arginine to ornithine and urea by host arginase supports Leishmania proliferation in macrophages. Studies using arginase-null mutants show that Leishmania-derived arginase plays an important role in disease pathogenesis. We investigated the role of parasite-derived arginase in secondary (memory) anti-Leishmania immunity in the resistant C57BL/6 mice. We found that C57BL/6 mice infected with arginase-deficient (arg(-)) L. major failed to completely resolve their lesion and maintained chronic pathology after 16 wk, a time when the lesion induced by wild-type L. major is completely resolved. This chronic disease was associated with impaired Ag-specific proliferation and IFN-γ production, a concomitant increase in programmed cell death-1 (PD-1) expression on CD4(+) T cells, and failure to induce protection against secondary L. major challenge. Treatment with anti-PD-1 mAb restored T cell proliferation and IFN-γ production in vitro and led to complete resolution of chronic lesion in arg(-) L. major-infected mice. These results show that infection with arg(-) L. major results in chronic disease due in part to PD-1-mediated clonal exhaustion of T cells, suggesting that parasite-derived arginase contributes to the overall quality of the host immune response and subsequent disease outcome in L. major-infected mice. They also indicate that persistent parasites alone do not regulate the quality of secondary anti-Leishmania immunity in mice and that the quality of the primary immune response may be playing a hitherto unrecognized dominant role in this process.
Collapse
Affiliation(s)
- Zhirong Mou
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba R3E 0T5, Canada
| | | | | | | | | | | | | | | |
Collapse
|
128
|
Carrión J, Abengozar MA, Fernández-Reyes M, Sánchez-Martín C, Rial E, Domínguez-Bernal G, González-Barroso MM. UCP2 deficiency helps to restrict the pathogenesis of experimental cutaneous and visceral leishmaniosis in mice. PLoS Negl Trop Dis 2013; 7:e2077. [PMID: 23437414 PMCID: PMC3578745 DOI: 10.1371/journal.pntd.0002077] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 01/11/2013] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Uncoupling protein 2 (UCP2) is a mitochondrial transporter that has been shown to lower the production of reactive oxygen species (ROS). Intracellular pathogens such as Leishmania upregulate UCP2 and thereby suppress ROS production in infected host tissues, allowing the multiplication of parasites within murine phagocytes. This makes host UCP2 and ROS production potential targets in the development of antileishmanial therapies. Here we explore how UCP2 affects the outcome of cutaneous leishmaniosis (CL) and visceral leishmaniosis (VL) in wild-type (WT) C57BL/6 mice and in C57BL/6 mice lacking the UCP2 gene (UCP2KO). METHODOLOGY AND FINDINGS To investigate the effects of host UCP2 deficiency on Leishmania infection, we evaluated parasite loads and cytokine production in target organs. Parasite loads were significantly lower in infected UCP2KO mice than in infected WT mice. We also found that UCP2KO mice produced significantly more interferon-γ (IFN-γ), IL-17 and IL-13 than WT mice (P<0.05), suggesting that UCP2KO mice are resistant to Leishmania infection. CONCLUSIONS In this way, UCP2KO mice were better able than their WT counterparts to overcome L. major and L. infantum infections. These findings suggest that upregulating host ROS levels, perhaps by inhibiting UPC2, may be an effective approach to preventing leishmaniosis.
Collapse
Affiliation(s)
- Javier Carrión
- Department of Animal Health, Veterinary Faculty, Complutense University of Madrid, Madrid, Spain.
| | | | | | | | | | | | | |
Collapse
|
129
|
Takele Y, Abebe T, Weldegebreal T, Hailu A, Hailu W, Hurissa Z, Ali J, Diro E, Sisay Y, Cloke T, Modolell M, Munder M, Tacchini-Cottier F, Müller I, Kropf P. Arginase activity in the blood of patients with visceral leishmaniasis and HIV infection. PLoS Negl Trop Dis 2013; 7:e1977. [PMID: 23349999 PMCID: PMC3547864 DOI: 10.1371/journal.pntd.0001977] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 11/05/2012] [Indexed: 11/21/2022] Open
Abstract
Background Visceral leishmaniasis is a parasitic disease associated with high mortality. The most important foci of visceral leishmaniasis in Ethiopia are in the Northwest and are predominantly associated with high rates of HIV co-infection. Co-infection of visceral leishmaniasis patients with HIV results in higher mortality, treatment failure and relapse. We have previously shown that arginase, an enzyme associated with immunosuppression, was increased in patients with visceral leishmaniasis and in HIV seropositive patients; further our results showed that high arginase activity is a marker of disease severity. Here, we tested the hypothesis that increased arginase activities associated with visceral leishmaniasis and HIV infections synergize in patients co-infected with both pathogens. Methodology/Principal Findings We recruited a cohort of patients with visceral leishmaniasis and a cohort of patients with visceral leishmaniasis and HIV infection from Gondar, Northwest Ethiopia, and recorded and compared their clinical data. Further, we measured the levels of arginase activity in the blood of these patients and identified the phenotype of arginase-expressing cells. Our results show that CD4+ T cell counts were significantly lower and the parasite load in the spleen was significantly higher in co-infected patients. Moreover, our results demonstrate that arginase activity was significantly higher in peripheral blood mononuclear cells and plasma of co-infected patients. Finally, we identified the cells-expressing arginase in the PBMCs as low-density granulocytes. Conclusion Our results suggest that increased arginase might contribute to the poor disease outcome characteristic of patients with visceral leishmaniasis and HIV co-infection. Visceral leishmaniasis (VL) in Ethiopia is caused by the parasite Leishmania donovani. This disease has one of the highest mortality rates: if left untreated, it is almost always fatal. VL belongs to the most neglected tropical diseases, affecting the poorest populations, for whom access to diagnosis and effective treatment are often not available. VL is worsened when the patients are co-infected with HIV. We have recently shown that patients with visceral leishmaniasis (VL patients) and HIV seropositive patients (HIV+ patients) have increased levels of an enzyme, arginase, in their blood. This enzyme has been shown to prevent cells of the immune system from mounting an efficient response and controlling the replication of the virus in HIV+ patients or the parasites in VL patients. In this study, we show that arginase is considerably higher in the blood of VL/HIV co-infected patients as compared to VL patients. Our results suggest that this abnormally high arginase might contribute to the poor prognosis associated to VL/HIV patients.
Collapse
Affiliation(s)
- Yegnasew Takele
- Gondar University Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Tamrat Abebe
- Department of Microbiology, Immunology, Parasitology and Pathology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Teklu Weldegebreal
- Gondar University Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Asrat Hailu
- Department of Microbiology, Immunology, Parasitology and Pathology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Workagegnehu Hailu
- College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Zewdu Hurissa
- Gondar University Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Jemal Ali
- Department of Microbiology, Immunology and Parasitology, School of Biomedical and Laboratory Sciences, University of Gondar, Gondar, Ethiopia
| | - Ermiyas Diro
- Gondar University Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Yifru Sisay
- College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Tom Cloke
- Department of Medicine, Section of Immunology, Imperial College London, London, United Kingdom
| | - Manuel Modolell
- Department of Cellular Immunology, Max-Planck-Institute for Immunobiology and Epigenetics, Freiburg, Germany
| | - Markus Munder
- Third Department of Medicine (Hematology, Oncology, and Pneumology), University Medical Center Mainz, Mainz, Germany
| | - Fabienne Tacchini-Cottier
- Department of Biochemistry, WHO Immunology Research and Training Center, University of Lausanne, Lausanne, Switzerland
| | - Ingrid Müller
- Department of Medicine, Section of Immunology, Imperial College London, London, United Kingdom
| | - Pascale Kropf
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
- * E-mail:
| |
Collapse
|
130
|
Abstract
Leishmaniasis is a vector-borne neglected tropical disease associated with a spectrum of clinical manifestations, ranging from self-healing cutaneous lesions to fatal visceral infections. Among the most important questions in Leishmania research is why some species like L. donovani infect visceral organs, whereas other species like L. major remain in the skin. The determinants of visceral leishmaniasis are still poorly understood, although genomic, immunologic, and animal models are beginning to provide important insight into this disease. In this review, we discuss the vector, host, and pathogen factors that mediate the development of visceral leishmaniasis. We examine the progression of the parasite from the initial site of sand fly bite to the visceral organs and its ability to survive there. The identification of visceral disease determinants is required to understand disease evolution, to understand visceral organ survival mechanisms, and potentially to develop better interventions for this largely neglected disease.
Collapse
|
131
|
Bhaumik P, St-Pierre G, Milot V, St-Pierre C, Sato S. Galectin-3 Facilitates Neutrophil Recruitment as an Innate Immune Response to a Parasitic Protozoa Cutaneous Infection. THE JOURNAL OF IMMUNOLOGY 2012; 190:630-40. [DOI: 10.4049/jimmunol.1103197] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
132
|
Characterization of a novel population of low-density granulocytes associated with disease severity in HIV-1 infection. PLoS One 2012; 7:e48939. [PMID: 23152825 PMCID: PMC3496742 DOI: 10.1371/journal.pone.0048939] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 10/02/2012] [Indexed: 11/19/2022] Open
Abstract
The mechanisms resulting in progressive immune dysfunction during the chronic phase of HIV infection are not fully understood. We have previously shown that arginase, an enzyme with potent immunosuppressive properties, is increased in HIV seropositive (HIV+) patients with low CD4+ T cell counts. Here we show that the cells expressing arginase in peripheral blood mononuclear cells of HIV+ patients are low-density granulocytes (LDGs) and that whereas these cells have a similar morphology to normal-density granulocyte, they are phenotypically different. Importantly, our results reveal that increased frequencies of LDGs correlate with disease severity in HIV+ patients.
Collapse
|
133
|
Serrat N, Pereira-Lopes S, Comalada M, Lloberas J, Celada A. Deacetylation of C/EBPβ is required for IL-4-induced arginase-1 expression in murine macrophages. Eur J Immunol 2012; 42:3028-37. [PMID: 22865229 DOI: 10.1002/eji.201242413] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 07/26/2012] [Accepted: 07/30/2012] [Indexed: 12/31/2022]
Abstract
The amount of arginine available at inflammatory loci is a limiting factor for the growth of several cells of the immune system. IL-4-induced activation of macrophages produced arginase-1, which converts arginine into ornithine, a precursor of polyamines and proline. Trichostatin A (TSA), a pan-inhibitor of histone deacetylases (HDACs), inhibited IL-4-induced arginase-1 expression. TSA showed promoter-specific effects on the IL-4-responsive genes. While TSA inhibited the expression of arginase-1, fizz1, and mrc1, other genes, such as ym,1 mgl1, and mgl2, were not affected. The inhibition of arginase-1 occurred at the transcriptional level with the inhibition of polymerase II binding to the promoter. IL-4 induced STAT6 phosphorylation and binding to DNA. These activities were not affected by TSA treatment. However, TSA inhibited C/EBPβ DNA binding. This inhibitor induced acetylation on lysine residues 215-216, which are critical for DNA binding. Finally, using macrophages from STAT6 KO mice we showed that STAT6 is required for the DNA binding of C/EBPβ. These results demonstrate that the acetylation/deacetylation balance strongly influences the expression of arginase-1, a gene of alternative activation of macrophages. These findings also provide a molecular mechanism to explain the control of gene expression through deacetylase activity.
Collapse
Affiliation(s)
- Neus Serrat
- Institute for Research in Biomedicine, Barcelona, Spain
| | | | | | | | | |
Collapse
|
134
|
Domínguez-Bernal G, Horcajo P, Orden JA, De La Fuente R, Herrero-Gil A, Ordóñez-Gutiérrez L, Carrión J. Mitigating an undesirable immune response of inherent susceptibility to cutaneous leishmaniosis in a mouse model: the role of the pathoantigenic HISA70 DNA vaccine. Vet Res 2012; 43:59. [PMID: 22876751 PMCID: PMC3503552 DOI: 10.1186/1297-9716-43-59] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 07/18/2012] [Indexed: 02/05/2023] Open
Abstract
Leishmania major is the major cause of cutaneous leishmaniosis (CL) outside of the Americas. In the present study we have cloned six Leishmania genes (H2A, H2B, H3, H4, A2 and HSP70) into the eukaryotic expression vector pCMVβ-m2a, resulting in pCMV-HISA70m2A, which encodes all six pathoantigenic proteins as a single polyprotein. This expression plasmid has been evaluated as a novel vaccine candidate in the BALB/c mouse model of CL. The DNA vaccine shifted the immune response normally induced by L. major infection away from a Th2-specific pathway to one of basal susceptibility. Immunization with pCMV-HISA70m2A dramatically reduced footpad lesions and lymph node parasite burdens relative to infected control mice. Complete absence of visceral parasite burden was observed in all 12 immunized animals but not in any of the 24 control mice. Moreover, vaccinated mice produced large amounts of IFN-γ, IL-17 and NO at 7 weeks post-infection (pi), and they showed lower arginase activity at the site of infection, lower IL-4 production and a weaker humoral immune response than infected control mice. Taken together, these results demonstrate the ability of the HISA70 vaccine to shift the murine immune response to L. major infection away from an undesirable, Th2-specific pathway to a less susceptible-like pathway involving Th1 and Th17 cytokine profiles.
Collapse
Affiliation(s)
- Gustavo Domínguez-Bernal
- Department of Animal Health, Veterinary Faculty, Complutense University of Madrid, 28040, Madrid, Spain.
| | | | | | | | | | | | | |
Collapse
|
135
|
Paul J, Karmakar S, De T. TLR-mediated distinct IFN-γ/IL-10 pattern induces protective immunity against murine visceral leishmaniasis. Eur J Immunol 2012; 42:2087-99. [DOI: 10.1002/eji.201242428] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Joydeep Paul
- Division of Infectious Disease and Immunology; Council of Scientific and Industrial Research; Indian Institute of Chemical Biology; Kolkata; India
| | - Subir Karmakar
- Division of Infectious Disease and Immunology; Council of Scientific and Industrial Research; Indian Institute of Chemical Biology; Kolkata; India
| | - Tripti De
- Division of Infectious Disease and Immunology; Council of Scientific and Industrial Research; Indian Institute of Chemical Biology; Kolkata; India
| |
Collapse
|
136
|
Ming X, Rajapakse AG, Yepuri G, Xiong Y, Carvas JM, Ruffieux J, Scerri I, Wu Z, Popp K, Li J, Sartori C, Scherrer U, Kwak BR, Montani J, Yang Z. Arginase II Promotes Macrophage Inflammatory Responses Through Mitochondrial Reactive Oxygen Species, Contributing to Insulin Resistance and Atherogenesis. J Am Heart Assoc 2012; 1:e000992. [PMID: 23130157 PMCID: PMC3487353 DOI: 10.1161/jaha.112.000992] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 06/08/2012] [Indexed: 12/29/2022]
Abstract
BACKGROUND Macrophage-mediated chronic inflammation is mechanistically linked to insulin resistance and atherosclerosis. Although arginase I is considered antiinflammatory, the role of arginase II (Arg-II) in macrophage function remains elusive. This study characterizes the role of Arg-II in macrophage inflammatory responses and its impact on obesity-linked type II diabetes mellitus and atherosclerosis. METHODS AND RESULTS In human monocytes, silencing Arg-II decreases the monocytes' adhesion to endothelial cells and their production of proinflammatory mediators stimulated by oxidized low-density lipoprotein or lipopolysaccharides, as evaluated by real-time quantitative reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay. Macrophages differentiated from bone marrow cells of Arg-II-deficient (Arg-II(-/-)) mice express lower levels of lipopolysaccharide-induced proinflammatory mediators than do macrophages of wild-type mice. Importantly, reintroducing Arg-II cDNA into Arg-II(-/-) macrophages restores the inflammatory responses, with concomitant enhancement of mitochondrial reactive oxygen species. Scavenging of reactive oxygen species by N-acetylcysteine prevents the Arg-II-mediated inflammatory responses. Moreover, high-fat diet-induced infiltration of macrophages in various organs and expression of proinflammatory cytokines in adipose tissue are blunted in Arg-II(-/-) mice. Accordingly, Arg-II(-/-) mice reveal lower fasting blood glucose and improved glucose tolerance and insulin sensitivity. Furthermore, apolipoprotein E (ApoE)-deficient mice with Arg-II deficiency (ApoE(-/-)Arg-II(-/-)) display reduced lesion size with characteristics of stable plaques, such as decreased macrophage inflammation and necrotic core. In vivo adoptive transfer experiments reveal that fewer donor ApoE(-/-)Arg-II(-/-) than ApoE(-/-)Arg-II(+/+) monocytes infiltrate into the plaque of ApoE(-/-)Arg-II(+/+) mice. Conversely, recipient ApoE(-/-)Arg-II(-/-) mice accumulate fewer donor monocytes than do recipient ApoE(-/-)Arg-II(+/+) animals. CONCLUSIONS Arg-II promotes macrophage proinflammatory responses through mitochondrial reactive oxygen species, contributing to insulin resistance and atherogenesis. Targeting Arg-II represents a potential therapeutic strategy in type II diabetes mellitus and atherosclerosis. (J Am Heart Assoc. 2012;1:e000992 doi: 10.1161/JAHA.112.000992.).
Collapse
Affiliation(s)
- Xiu‐Fen Ming
- From the Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, Faculty of Science, University of Fribourg, Switzerland (X.-F.M.,
A.G.R., G.Y., Y.X., J.M.C., J.R., I.S., Z.W., K.P., J.-P.M., Z.Y.)
| | - Angana G. Rajapakse
- From the Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, Faculty of Science, University of Fribourg, Switzerland (X.-F.M.,
A.G.R., G.Y., Y.X., J.M.C., J.R., I.S., Z.W., K.P., J.-P.M., Z.Y.)
| | - Gautham Yepuri
- From the Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, Faculty of Science, University of Fribourg, Switzerland (X.-F.M.,
A.G.R., G.Y., Y.X., J.M.C., J.R., I.S., Z.W., K.P., J.-P.M., Z.Y.)
| | - Yuyan Xiong
- From the Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, Faculty of Science, University of Fribourg, Switzerland (X.-F.M.,
A.G.R., G.Y., Y.X., J.M.C., J.R., I.S., Z.W., K.P., J.-P.M., Z.Y.)
| | - João M. Carvas
- From the Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, Faculty of Science, University of Fribourg, Switzerland (X.-F.M.,
A.G.R., G.Y., Y.X., J.M.C., J.R., I.S., Z.W., K.P., J.-P.M., Z.Y.)
| | - Jean Ruffieux
- From the Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, Faculty of Science, University of Fribourg, Switzerland (X.-F.M.,
A.G.R., G.Y., Y.X., J.M.C., J.R., I.S., Z.W., K.P., J.-P.M., Z.Y.)
| | - Isabelle Scerri
- From the Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, Faculty of Science, University of Fribourg, Switzerland (X.-F.M.,
A.G.R., G.Y., Y.X., J.M.C., J.R., I.S., Z.W., K.P., J.-P.M., Z.Y.)
| | - Zongsong Wu
- From the Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, Faculty of Science, University of Fribourg, Switzerland (X.-F.M.,
A.G.R., G.Y., Y.X., J.M.C., J.R., I.S., Z.W., K.P., J.-P.M., Z.Y.)
| | - Katja Popp
- From the Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, Faculty of Science, University of Fribourg, Switzerland (X.-F.M.,
A.G.R., G.Y., Y.X., J.M.C., J.R., I.S., Z.W., K.P., J.-P.M., Z.Y.)
| | - Jianhui Li
- Department of Intensive Care Medicine, University Hospital Center and Faculty of Biology and Medicine, Lausanne, Switzerland (J.L.)
- Dr Li is currently affiliated with the Department of Hepatobiliary and Pancreatic Surgery and Centre of Organ Transplantation, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China (J.L.)
| | - Claudio Sartori
- Department of Internal Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland (C.S.)
| | - Urs Scherrer
- Department of Cardiology, University Hospital, Bern, Switzerland (U.S.)
- Facultad de Ciencias, Departamento de Biología, Universidad de Tarapacá, Arica, Chile (U.S.)
| | - Brenda R. Kwak
- Department of Pathology and Immunology, Department of Internal Medicine–Cardiology, University of Geneva, Switzerland (B.R.K.)
| | - Jean‐Pierre Montani
- From the Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, Faculty of Science, University of Fribourg, Switzerland (X.-F.M.,
A.G.R., G.Y., Y.X., J.M.C., J.R., I.S., Z.W., K.P., J.-P.M., Z.Y.)
| | - Zhihong Yang
- From the Laboratory of Vascular Biology, Department of Medicine, Division of Physiology, Faculty of Science, University of Fribourg, Switzerland (X.-F.M.,
A.G.R., G.Y., Y.X., J.M.C., J.R., I.S., Z.W., K.P., J.-P.M., Z.Y.)
| |
Collapse
|
137
|
Aguiar DN, Silva MM, Parreira WV, Tome FD, Batista LF, Gomes CM, Oliveira MA. Electroacupuncture at the ST36 acupoint increases interleukin-4 responsiveness in macrophages, generation of alternatively activated macrophages and susceptibility to Leishmania major infection. Chin Med 2012; 7:17. [PMID: 22838729 PMCID: PMC3444353 DOI: 10.1186/1749-8546-7-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 07/12/2012] [Indexed: 12/11/2022] Open
Abstract
Background Electroacupuncture (EA) has been used to treat inflammatory diseases. Alternatively activated macrophages (AAMo) stimulated by cytokines such as interleukin (IL)-4, IL-10 and IL-13 are anti-inflammatory and mildly microbicidal. This study aimed to evaluate whether EA at the Zusanli acupoint (ST36) would change the profile of healthy murine macrophages, particularly the generation of AAMo and susceptibility to Leishmania major infection. Methods BALB/c mice were treated with EA (15/30 Hz) at the ST36 acupoint for 20 min/d for 5 d. After the final EA session, the mice were euthanized and their peritoneal cells were harvested and counted for determination of arginase activity, nitric oxide (NO) production and microbicidal activity after culture in the presence or absence of IL-4, interferon-γ (IFNγ) or lipopolysaccharide (LPS) or both IFNγ and LPS. Twelve mice were infected with L. major promastigotes into the footpads after the final EA session and the infection course was monitored. Results Peritoneal cells freshly obtained from EA-treated mice had similar arginase and microbicidal activities to cells from sham-treated mice. After culture with IL-4, cells from EA-treated mice exhibited significant increases in the arginase activity (sham: 58 ± 11.3 vs. EA: 80.7 ± 4.6%, P = 0.025) and number of parasites/infected cell (sham: 2.5 ± 0.4 vs. EA: 4.3 ± 0.8 cells, P = 0.007). The NO production was lower in cells from EA-treated mice cultured in the presence of a combination of IFNγ and LPS (sham: 31.6 ± 6.5 vs. EA: 22.3 ± 2.1 μM, P = 0.025). The lesion size in mice infected with L. major promastigotes was larger in EA-treated mice (sham: 3.26 ± 0.29 vs. EA: 2.23 ± 0.4 mm, P = 0.039). Conclusion EA at the ST36 acupoint increases IL-4 responsiveness in macrophages, Generation of AAMo and susceptibility to L. major infection
Collapse
Affiliation(s)
- Danillo N Aguiar
- Department of Microbiology, Immunology, Parasitology and Pathology, Tropical Pathology and Public Health Institute, Federal University of Goiás, Goiânia, Goiás, Brazil.
| | | | | | | | | | | | | |
Collapse
|
138
|
Lamour SD, Choi BS, Keun HC, Müller I, Saric J. Metabolic characterization of Leishmania major infection in activated and nonactivated macrophages. J Proteome Res 2012; 11:4211-22. [PMID: 22724526 PMCID: PMC3411194 DOI: 10.1021/pr3003358] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
![]()
Infection with Leishmania spp. can lead
to a range
of symptoms in the affected individual, depending on underlying immune-metabolic
processes. The macrophage activation state hereby plays a key role.
Whereas the l-arginine pathway has been described in detail
as the main biochemical process responsible for either nitric oxide
mediated parasite killing (classical activation) or amplification
of parasite replication (alternative activation), we were interested
in a wider characterization of metabolic events in vitro. We therefore assessed cell growth medium, parasite extract, and
intra- and extracellular metabolome of activated and nonactivated
macrophages, in presence and absence of Leishmania major. A metabolic profiling approach was applied combining 1H NMR spectroscopy with multi- and univariate data treatment. Metabolic
changes were observed along both conditional axes, that is, infection
state and macrophage activation, whereby significantly higher levels
of potential parasite end products were found in parasite exposed
samples including succinate, acetate, and alanine, compared to uninfected
macrophages. The different macrophage activation states were mainly
discriminated by varying glucose consumption. The presented profiling
approach allowed us to obtain a metabolic snapshot of the individual
biological compartments in the assessed macrophage culture experiments
and represents a valuable read out system for further multiple compartment in vitro studies.
Collapse
Affiliation(s)
- Sabrina D Lamour
- Biomolecular Medicine, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, United Kingdom
| | | | | | | | | |
Collapse
|
139
|
Rogers ME. The role of leishmania proteophosphoglycans in sand fly transmission and infection of the Mammalian host. Front Microbiol 2012; 3:223. [PMID: 22754550 PMCID: PMC3384971 DOI: 10.3389/fmicb.2012.00223] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 05/31/2012] [Indexed: 11/17/2022] Open
Abstract
Leishmania are transmitted by the bite of their sand fly vector and this has a significant influence on the virulence of the resulting infection. From our studies into the interaction between parasite, vector, and host we have uncovered an important missing ingredient during Leishmania transmission. Leishmania actively adapt their sand fly hosts into efficient vectors by secreting Promastigote Secretory Gel (PSG), a proteophosphoglycan (PPG)-rich, mucin-like gel which accumulates in sand fly gut and mouthparts. This has the effect of blocking the fly, such that during bloodfeeding both parasites and gel are co-transmitted in an act of regurgitation. We are discovering that this has further implications for the mammalian infection, again, in favor of the parasite. Experimentally, PSG exacerbates cutaneous and visceral leishmaniasis and can promote the chronicity of Leishmania infection, even in mouse strains normally capable of controlling leishmaniasis. The underlying mechanism of PSG's action is a major focus of our ongoing work. This review aims to synthesize what is known about the role and action of PSG and its constituent proteophosphoglycans, for parasite colonization of the sand fly, transmission, and mammalian infection. Lastly, we discuss potential exploitation of this important vector-transmitted product and future avenues of research.
Collapse
Affiliation(s)
- Matthew E. Rogers
- Faculty of Infectious Tropical Diseases, Department of Immunology and Infection, London School of Hygiene and Tropical MedicineLondon, UK
| |
Collapse
|
140
|
Badaut J, Copin JC, Fukuda AM, Gasche Y, Schaller K, da Silva RF. Increase of arginase activity in old apolipoprotein-E deficient mice under Western diet associated with changes in neurovascular unit. J Neuroinflammation 2012; 9:132. [PMID: 22709928 PMCID: PMC3419627 DOI: 10.1186/1742-2094-9-132] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 06/18/2012] [Indexed: 12/19/2022] Open
Abstract
Aging and atherosclerosis are well-recognized risk factors for cardiac and neurovascular diseases. The Apolipoprotein E deficient (ApoE-/-) mouse on a high-fat diet is a classical model of atherosclerosis, characterized by the presence of atherosclerotic plaques in extracranial vessels but not in cerebral arteries. Increase in arginase activity was shown to participate in vascular dysfunction in the peripheral arteries of atherosclerotic mice by changing the level of nitric oxide (NO). NO plays a key role in the physiological functions of the neurovascular unit (NVU). However, the regulation of arginase expression and activity in the brain was never investigated in association with changes in the NVU, ApoE deficiency and high fat diet.Fourteen-month-old ApoE-/- mice on high-fat diet exhibited deposition of lipids in the NVU, impairment of blood-brain barrier properties, astrogliosis and an increase of aquaporin 4 staining. In association with these changes, brain arginase activity was significantly increased in the old ApoE-/- mice as compared to old wild type mice, with an increase in the level of arginase type I in the blood vessels.In conclusion, aging in this classical mouse model of atherosclerosis induces an increase in the level and activity of arginase I that may impair NO synthesis and contribute to changes in the NVU leading to blood-brain barrier leakage and inflammation.
Collapse
Affiliation(s)
- Jérôme Badaut
- Departments of Pediatrics and Physiology, Linda University School of Medicine, Coleman Pavilion, Room A1120, 11175 Campus Street, Loma Linda, CA 92354, USA.
| | | | | | | | | | | |
Collapse
|
141
|
Biological Roles of Liver X Receptors in Immune Cells. Arch Immunol Ther Exp (Warsz) 2012; 60:235-49. [DOI: 10.1007/s00005-012-0179-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 03/16/2012] [Indexed: 12/17/2022]
|
142
|
Liu D, Uzonna JE. The early interaction of Leishmania with macrophages and dendritic cells and its influence on the host immune response. Front Cell Infect Microbiol 2012; 2:83. [PMID: 22919674 PMCID: PMC3417671 DOI: 10.3389/fcimb.2012.00083] [Citation(s) in RCA: 220] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 05/27/2012] [Indexed: 01/26/2023] Open
Abstract
The complicated interactions between Leishmania and the host antigen-presenting cells (APCs) have fundamental effects on the final outcome of the disease. Two major APCs, macrophages and dendritic cells (DCs), play critical roles in mediating resistance and susceptibility during Leishmania infection. Macrophages are the primary resident cell for Leishmania: they phagocytose and permit parasite proliferation. However, these cells are also the major effector cells to eliminate infection. The effective clearance of parasites by macrophages depends on activation of appropriate immune response, which is usually initiated by DCs. Here, we review the early interaction of APCs with Leishmania parasites and how these interactions profoundly impact on the ensuing adaptive immune response. We also discuss how the current knowledge will allow further refinement of our understanding of the interplay between Leishmania and its hosts that leads to resistance or susceptibility.
Collapse
Affiliation(s)
- Dong Liu
- Department of Immunology, University of Manitoba, Winnipeg MB, Canada
| | | |
Collapse
|
143
|
Local increase of arginase activity in lesions of patients with cutaneous leishmaniasis in Ethiopia. PLoS Negl Trop Dis 2012; 6:e1684. [PMID: 22720104 PMCID: PMC3373636 DOI: 10.1371/journal.pntd.0001684] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 04/27/2012] [Indexed: 11/28/2022] Open
Abstract
Background Cutaneous leishmaniasis is a vector-borne disease that is in Ethiopia mainly caused by the parasite Leishmania aethiopica. This neglected tropical disease is common in rural areas and causes serious morbidity. Persistent nonhealing cutaneous leishmaniasis has been associated with poor T cell mediated responses; however, the underlying mechanisms are not well understood. Methodology/Principal Findings We have recently shown in an experimental model of cutaneous leishmaniasis that arginase-induced L-arginine metabolism suppresses antigen-specific T cell responses at the site of pathology, but not in the periphery. To test whether these results translate to human disease, we recruited patients presenting with localized lesions of cutaneous leishmaniasis and assessed the levels of arginase activity in cells isolated from peripheral blood and from skin biopsies. Arginase activity was similar in peripheral blood mononuclear cells (PBMCs) from patients and healthy controls. In sharp contrast, arginase activity was significantly increased in lesion biopsies of patients with localized cutaneous leishmaniasis as compared with controls. Furthermore, we found that the expression levels of CD3ζ, CD4 and CD8 molecules were considerably lower at the site of pathology as compared to those observed in paired PBMCs. Conclusion Our results suggest that increased arginase in lesions of patients with cutaneous leishmaniasis might play a role in the pathogenesis of the disease by impairing T cell effector functions. The leishmaniases are a complex of diseases caused by Leishmania parasites. Currently, the diseases affect an estimated 12 million people in 88 countries, and approximately 350 million more people are at risk. The leishmaniases belong to the most neglected tropical diseases, affecting the poorest populations, for whom access to diagnosis and effective treatment are often not available. Leishmania parasites infect cells of the immune system called macrophages, which have the capacity to eliminate the intracellular parasites when they receive the appropriate signals from other cells of the immune system. In nonhealing persistent leishmaniasis, lymphocytes are unable to transmit the signals to macrophages required to kill the intracellular parasites. The local upregulation of the enzyme arginase has been shown to impair lymphocyte effector functions at the site of pathology. In this study, we tested the activity of this enzyme in skin lesions of patients presenting with localized cutaneous leishmaniasis. Our results show that arginase is highly upregulated in these lesions. This increase in arginase activity coincides with lower expression of a signalling molecule in lymphocytes, which is essential for efficient activation of these cells. These results suggest that increased arginase expression in the localized cutaneous lesions might contribute to persistent disease in patients presenting with cutaneous leishmaniasis.
Collapse
|
144
|
Leishmania amazonensis arginase compartmentalization in the glycosome is important for parasite infectivity. PLoS One 2012; 7:e34022. [PMID: 22479507 PMCID: PMC3316525 DOI: 10.1371/journal.pone.0034022] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 02/20/2012] [Indexed: 11/19/2022] Open
Abstract
In Leishmania, de novo polyamine synthesis is initiated by the cleavage of L-arginine to urea and L-ornithine by the action of arginase (ARG, E.C. 3.5.3.1). Previous studies in L. major and L. mexicana showed that ARG is essential for in vitro growth in the absence of polyamines and needed for full infectivity in animal infections. The ARG protein is normally found within the parasite glycosome, and here we examined whether this localization is required for survival and infectivity. First, the localization of L. amazonensis ARG in the glycosome was confirmed in both the promastigote and amastigote stages. As in other species, arg− L. amazonensis required putrescine for growth and presented an attenuated infectivity. Restoration of a wild type ARG to the arg− mutant restored ARG expression, growth and infectivity. In contrast, restoration of a cytosol-targeted ARG lacking the glycosomal SKL targeting sequence (argΔSKL) restored growth but failed to restore infectivity. Further study showed that the ARGΔSKL protein was found in the cytosol as expected, but at very low levels. Our results indicate that the proper compartmentalization of L. amazonensis arginase in the glycosome is important for enzyme activity and optimal infectivity. Our conjecture is that parasite arginase participates in a complex equilibrium that defines the fate of L-arginine and that its proper subcellular location may be essential for this physiological orchestration.
Collapse
|
145
|
Ghosh J. Role of Nitric Oxide in Salmonella Infection. Indian J Clin Biochem 2012; 27:306-8. [PMID: 26405393 DOI: 10.1007/s12291-012-0187-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Accepted: 01/15/2012] [Indexed: 11/29/2022]
Abstract
Nitric oxide (NO) derivative of l-arginine is an important signaling molecule that mediates a variety of essential physiological processes including vasodilation neurotransmission, and host cell defense. Many types of cells produce NO e.g., smooth muscle cell, endothelial cell, and leukocytes. Host defense functions are known for many bacterial and parasitic infections. In the present study we estimated the levels of serum NO in cases of salmonellosis and in controls. The nitric oxide was estimated by cadmium reduction method, Griess reaction. We observed that in controls the level of NO was (22 ± 2.06) μmol/l and in cases the level was (137.49 ± 29.84) μmol/l. The level of NO was significantly higher than controls (p < 0.001). The raised level of NO could be accounted for by host response to the infection. The host rapidly expresses iNOS, which in turn produces an excess amount of NO. Its cytotoxic effect is by its reactive nitrogen oxide derivative e.g., peroxynitrite. Apart from this it also has anti apoptotic functions. In future one can do follow up study of typhoid cases by bacterial culture.
Collapse
Affiliation(s)
- Joya Ghosh
- Department of Biochemistry, ESI-PGIMSR, Joka, Diamond Harbour Road, Kolkata, 700 104 India
| |
Collapse
|
146
|
Van den Bossche J, Lamers WH, Koehler ES, Geuns JMC, Alhonen L, Uimari A, Pirnes-Karhu S, Van Overmeire E, Morias Y, Brys L, Vereecke L, De Baetselier P, Van Ginderachter JA. Pivotal Advance: Arginase-1-independent polyamine production stimulates the expression of IL-4-induced alternatively activated macrophage markers while inhibiting LPS-induced expression of inflammatory genes. J Leukoc Biol 2012; 91:685-99. [PMID: 22416259 DOI: 10.1189/jlb.0911453] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In macrophages, basal polyamine (putrescine, spermidine, and spermine) levels are relatively low but are increased upon IL-4 stimulation. This Th2 cytokine induces Arg1 activity, which converts arginine into ornithine, and ornithine can be decarboxylated by ODC to produce putrescine, which is further converted into spermidine and spermine. Recently, we proposed polyamines as novel agents in IL-4-dependent E-cadherin regulation in AAMs. Here, we demonstrate for the first time that several, but not all, AAM markers depend on polyamines for their IL-4-induced gene and protein expression and that polyamine dependency of genes relies on the macrophage type. Remarkably, Arg1-deficient macrophages display rather enhanced IL-4-induced polyamine production, suggesting that an Arg1-independent polyamine synthesis pathway may operate in macrophages. On the other side of the macrophage activation spectrum, LPS-induced expression of several proinflammatory genes was increased significantly in polyamine-depleted CAMs. Overall, we propose Arg1 independently produced polyamines as novel regulators of the inflammatory status of the macrophage. Indeed, whereas polyamines are needed for IL-4-induced expression of several AAM mediators, they inhibit the LPS-mediated expression of proinflammatory genes in CAMs.
Collapse
Affiliation(s)
- Jan Van den Bossche
- Myeloid Cell Immunology Lab, VIB-Vrije Universiteit Brussel, Building E, Level 8, Pleinlaan 2, B-1050, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Lacerda DI, Cysne-Finkelstein L, Nunes MP, De-Luca PM, Genestra MDS, Leon LLP, Berrêdo-Pinho M, Mendonça-Lima L, Matos DCDS, Medeiros MA, Mendonça SCFD. Kinetoplastid membrane protein-11 exacerbates infection with Leishmania amazonensis in murine macrophages. Mem Inst Oswaldo Cruz 2012; 107:238-45. [DOI: 10.1590/s0074-02762012000200014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Accepted: 12/07/2011] [Indexed: 11/22/2022] Open
|
148
|
The leishmanicidal flavonols quercetin and quercitrin target Leishmania (Leishmania) amazonensis arginase. Exp Parasitol 2012; 130:183-8. [DOI: 10.1016/j.exppara.2012.01.015] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 11/24/2011] [Accepted: 01/19/2012] [Indexed: 01/26/2023]
|
149
|
Role of trypanosomatid's arginase in polyamine biosynthesis and pathogenesis. Mol Biochem Parasitol 2012; 181:85-93. [DOI: 10.1016/j.molbiopara.2011.10.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 10/12/2011] [Accepted: 10/12/2011] [Indexed: 01/08/2023]
|
150
|
Osorio EY, Zhao W, Espitia C, Saldarriaga O, Hawel L, Byus CV, Travi BL, Melby PC. Progressive visceral leishmaniasis is driven by dominant parasite-induced STAT6 activation and STAT6-dependent host arginase 1 expression. PLoS Pathog 2012; 8:e1002417. [PMID: 22275864 PMCID: PMC3261917 DOI: 10.1371/journal.ppat.1002417] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 10/19/2011] [Indexed: 01/11/2023] Open
Abstract
The clinicopathological features of the hamster model of visceral leishmaniasis (VL) closely mimic active human disease. Studies in humans and hamsters indicate that the inability to control parasite replication in VL could be related to ineffective classical macrophage activation. Therefore, we hypothesized that the pathogenesis of VL might be driven by a program of alternative macrophage activation. Indeed, the infected hamster spleen showed low NOS2 but high arg1 enzyme activity and protein and mRNA expression (p<0.001) and increased polyamine synthesis (p<0.05). Increased arginase activity was also evident in macrophages isolated from the spleens of infected hamsters (p<0.05), and arg1 expression was induced by L. donovani in primary hamster peritoneal macrophages (p<0.001) and fibroblasts (p<0.01), and in a hamster fibroblast cell line (p<0.05), without synthesis of endogenous IL-4 or IL-13 or exposure to exogenous cytokines. miRNAi-mediated selective knockdown of hamster arginase 1 (arg1) in BHK cells led to increased generation of nitric oxide and reduced parasite burden (p<0.005). Since many of the genes involved in alternative macrophage activation are regulated by Signal Transducer and Activator of Transcription-6 (STAT6), and because the parasite-induced expression of arg1 occurred in the absence of exogenous IL-4, we considered the possibility that L. donovani was directly activating STAT6. Indeed, exposure of hamster fibroblasts or macrophages to L. donovani resulted in dose-dependent STAT6 activation, even without the addition of exogenous cytokines. Knockdown of hamster STAT6 in BHK cells with miRNAi resulted in reduced arg1 mRNA expression and enhanced control of parasite replication (p<0.0001). Collectively these data indicate that L. donovani infection induces macrophage STAT6 activation and STAT6-dependent arg1 expression, which do not require but are amplified by type 2 cytokines, and which contribute to impaired control of infection. Visceral leishmaniasis (VL), caused by the intracellular protozoan Leishmania donovani, is a progressive, potentially fatal infection found in many resource-poor regions of the world. We initiated these studies of an experimental model of VL to better understand the molecular and cellular determinants underlying this disease. We found that host macrophages or fibroblasts, when infected with Leishmania donovani or exposed to products secreted by the parasite, are permissive to infection because they fail to metabolize arginine to generate nitric oxide, the effector molecule needed to kill the intracellular parasites. Instead, the infected host cells are activated in a way that leads to the expression of arginase, an enzyme that metabolizes arginine to produce polyamines, which support parasite growth. This detrimental activation pathway was dependent on the parasite-induced activation of the transcription factor STAT6, but contrary to the previously accepted paradigm, did not require (but was amplified by) the presence of polarized Th2 cells or type 2 cytokines. Knockdown of host arginase or STAT6 enhanced control of the infection, indicating that this activation pathway has a critical role in the pathogenesis of the disease. Interventions designed to inhibit the STAT6-arginase-polyamine pathway could help in the treatment or prevention of VL.
Collapse
Affiliation(s)
- E. Yaneth Osorio
- Research Service, Department of Veterans Affairs Medical Center, South Texas Veterans Health Care System, San Antonio, Texas, United States of America
- Department of Medicine, The University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Weiguo Zhao
- Research Service, Department of Veterans Affairs Medical Center, South Texas Veterans Health Care System, San Antonio, Texas, United States of America
- Department of Medicine, The University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Claudia Espitia
- Research Service, Department of Veterans Affairs Medical Center, South Texas Veterans Health Care System, San Antonio, Texas, United States of America
- Department of Medicine, The University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Omar Saldarriaga
- Research Service, Department of Veterans Affairs Medical Center, South Texas Veterans Health Care System, San Antonio, Texas, United States of America
- Department of Microbiology and Immunology, The University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Leo Hawel
- Division of Biomedical Sciences, University of California, Riverside, California, United States of America
| | - Craig V. Byus
- Division of Biomedical Sciences, University of California, Riverside, California, United States of America
| | - Bruno L. Travi
- Research Service, Department of Veterans Affairs Medical Center, South Texas Veterans Health Care System, San Antonio, Texas, United States of America
- Department of Medicine, The University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Peter C. Melby
- Research Service, Department of Veterans Affairs Medical Center, South Texas Veterans Health Care System, San Antonio, Texas, United States of America
- Department of Medicine, The University of Texas Health Science Center, San Antonio, Texas, United States of America
- Department of Microbiology and Immunology, The University of Texas Health Science Center, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|