1
|
Sanchez S, Chimenti MS, Lu Y, Sagues E, Gudino A, Dier C, Hasan D, Samaniego EA. Modulation of the Immunological Milieu in Acute Aneurysmal Subarachnoid Hemorrhage: The Potential Role of Monocytes Through CXCL10 Secretion. Transl Stroke Res 2025; 16:88-95. [PMID: 38780865 DOI: 10.1007/s12975-024-01259-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Abstract
Emerging evidence indicates that aneurysmal subarachnoid hemorrhage (aSAH) elicits a response from both innate and adaptive immune systems. An upregulation of CD8 + CD161 + cells has been observed in the cerebrospinal fluid (CSF) after aSAH, yet the precise role of these cells in the context of aSAH is unkown. CSF samples from patients with aSAH and non-aneurysmal SAH (naSAH) were analyzed. Single-cell RNA sequencing (scRNAseq) was performed on CD8 + CD161 + sorted samples from aSAH patients. Cell populations were identified using "clustering." Gene expression levels of ten previously described genes involved in inflammation were quantified from aSAH and naSAH samples using RT-qPCR. The study focused on the following genes: CCL5, CCL7, APOE, SPP1, CXCL8, CXCL10, HMOX1, LTB, MAL, and HLA-DRB1. Gene clustering analysis revealed that monocytes, NK cells, and T cells expressed CD8 + CD161 + in the CSF of patients with aSAH. In comparison to naSAH samples, aSAH samples exhibited higher mRNA levels of CXCL10 (median, IQR = 90, 16-149 vs. 0.5, 0-6.75, p = 0.02). A trend towards higher HMOX1 levels was also observed in aSAH (median, IQR = 12.6, 9-17.6 vs. 2.55, 1.68-5.7, p = 0.076). Specifically, CXCL10 and HMOX1 were expressed by the monocyte subpopulation. Monocytes, NK cells, and T cells can potentially express CD8 + CD161 + in patients with aSAH. Notably, monocytes show high levels of CXCL10. The elevated expression of CXCL10 in aSAH compared to naSAH indicates its potential significance as a target for future studies.
Collapse
Affiliation(s)
| | | | - Yongjun Lu
- Department of Neurology, University of Iowa, Iowa, IA, USA
| | - Elena Sagues
- Department of Neurology, University of Iowa, Iowa, IA, USA
| | - Andres Gudino
- Department of Neurology, University of Iowa, Iowa, IA, USA
| | - Carlos Dier
- Department of Neurology, University of Iowa, Iowa, IA, USA
| | - David Hasan
- Department of Neurosurgery, Duke University, Durham, NC, USA
| | - Edgar A Samaniego
- Department of Neurology, University of Iowa, Iowa, IA, USA.
- Department of Neurosurgery, University of Iowa, Iowa, IA, USA.
- Department of Radiology, University of Iowa, Iowa, IA, USA.
| |
Collapse
|
2
|
Huang CH, Laurent-Rolle M, Grove TL, Hsu JCC. Interferon-Stimulated Genes and Immune Metabolites as Broad-Spectrum Biomarkers for Viral Infections. Viruses 2025; 17:132. [PMID: 39861921 PMCID: PMC11768885 DOI: 10.3390/v17010132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
The type I interferon (IFN-I) response is a critical component of the immune defense against various viral pathogens, triggering the expression of hundreds of interferon-stimulated genes (ISGs). These ISGs encode proteins with diverse antiviral functions, targeting various stages of viral replication and restricting infection spread. Beyond their antiviral functions, ISGs and associated immune metabolites have emerged as promising broad-spectrum biomarkers that can differentiate viral infections from other conditions. This review provides an overview of the diagnostic potential of ISGs at transcript and protein levels, as well as their immune metabolites. We focus on their clinical applications and the sensitivity and specificity of these biomarkers through receiver operating characteristic (ROC) analysis. We highlight the need for further research to facilitate the effective translation of these biomarkers into clinical practice.
Collapse
Affiliation(s)
- Chien-Hsin Huang
- Center for Virus-Host-Innate-Immunity, Institute for Infectious and Inflammatory Diseases, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA;
| | - Maudry Laurent-Rolle
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA;
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Tyler L. Grove
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Jack Chun-Chieh Hsu
- Center for Virus-Host-Innate-Immunity, Institute for Infectious and Inflammatory Diseases, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA;
- Department of Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| |
Collapse
|
3
|
Upadhyay S, Kumar S, Singh VK, Tiwari R, Kumar A, Sundar S, Kumar R. Chemokines Signature and T Cell Dynamics in Leishmaniasis: Molecular insight and therapeutic application. Expert Rev Mol Med 2024; 27:1-55. [PMID: 39587036 PMCID: PMC11707835 DOI: 10.1017/erm.2024.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 09/04/2024] [Accepted: 10/28/2024] [Indexed: 11/27/2024]
Abstract
Leishmaniasis, caused by obligate intracellular Leishmania parasites, poses a significant global health burden. The control of Leishmania infection relies on an effective T cell-dependent immune response; however, various factors impede the host’s ability to mount a successful defence. Alterations in the chemokine profile, responsible for cell trafficking to the infection site, can disrupt optimal immune responses and influence the outcome of pathogenesis by facilitating parasite persistence. This review aims to emphasize the significance of the chemokine system in T cell responses and to summarize the current knowledge on the dysregulation of chemokines and their receptors associated with different subsets of T lymphocytes during Leishmaniasis. A comprehensive understanding of the dynamic nature of the chemokine system during Leishmaniasis is crucial for the development of successful immunotherapeutic approaches.
Collapse
Affiliation(s)
- Shreya Upadhyay
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shashi Kumar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vishal Kumar Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rahul Tiwari
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Awnish Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rajiv Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
4
|
Lafleur A, Daffis S, Mowbray C, Arana B. Immunotherapeutic Strategies as Potential Treatment Options for Cutaneous Leishmaniasis. Vaccines (Basel) 2024; 12:1179. [PMID: 39460345 PMCID: PMC11511131 DOI: 10.3390/vaccines12101179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Cutaneous leishmaniasis (CL), caused by protozoan parasites of the Leishmania genus, is prevalent in tropical and subtropical regions, with important morbidity, particularly in low- to middle-income countries. Current systemic treatments, including pentavalent antimonials and miltefosine, are associated with significant toxicity, reduced efficacy, and are frequently ineffective in cases of severe or chronic CL. Immunotherapies leverage the immune system to combat microbial infection and offer a promising adjunct or alternative approach to the current standard of care for CL. However, the heterogeneous clinical presentation of CL, which is dependent on parasite species and host immunity, may require informed clinical intervention with immunotherapies. This review explores the clinical and immunological characteristics of CL, emphasising the current landscape of immunotherapies in in vivo models and clinical studies. Such immune-based interventions aim to modulate immune responses against Leishmania, with additive therapeutic effects enabling the efficacy of lower drug doses and decreasing the associated toxicity. Understanding the mechanisms that underlie immunotherapy for CL provides critical insights into developing safer and more effective treatments for this neglected tropical disease. Identifying suitable therapeutic candidates and establishing their safety and efficacy are essential steps in this process. However, the feasibility and utility of these treatments in resource-limited settings must also be considered, taking into account factors such as cost of production, temperature stability, and overall patient access.
Collapse
Affiliation(s)
- Andrea Lafleur
- Doctoral Training Centre, University of Oxford, Oxford OX1 3NP, UK
| | - Stephane Daffis
- Drugs for Neglected Diseases initiative (DNDi), 1202 Geneva, Switzerland; (S.D.)
| | - Charles Mowbray
- Drugs for Neglected Diseases initiative (DNDi), 1202 Geneva, Switzerland; (S.D.)
| | - Byron Arana
- Drugs for Neglected Diseases initiative (DNDi), 1202 Geneva, Switzerland; (S.D.)
| |
Collapse
|
5
|
Yizengaw E, Takele Y, Franssen S, Gashaw B, Yimer M, Adem E, Nibret E, Yismaw G, Cruz Cervera E, Ejigu K, Tamiru D, Munshea A, Müller I, Weller R, Cotton JA, Kropf P. Investigation of parasite genetic variation and systemic immune responses in patients presenting with different clinical presentations of cutaneous leishmaniasis caused by Leishmania aethiopica. Infect Dis Poverty 2024; 13:76. [PMID: 39415297 PMCID: PMC11484111 DOI: 10.1186/s40249-024-01244-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/17/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Cutaneous leishmaniasis (CL) is a neglected tropical skin disease, caused by the protozoan parasite Leishmania. In Ethiopia, CL is mainly caused by Leishmania aethiopica and can present in different clinical forms. The aim of this study was to assess whether these different forms are associated with differences in parasite genetic and host systemic immune signatures. METHODS Here we analysed the whole genome sequence data for 48 clinical parasite isolates and the systemic immune signature from a cohort of CL patients, who were recruited in Nefas Mewcha, Northern Ethiopia, from January 2019 to January 2022. RESULTS Our results show that parasites from CL cases with different presentations in a single Ethiopian setting are from the same genetic population based on a permutation test of genome-wide similarity. Furthermore, a logistic regression test for genome wide association did not identify any individual genetic variants significantly associated with disease presentation. We also measured plasma chemokine and cytokine levels of 129 CL patients presenting with different forms of CL. None of the chemokine [eotaxin, eotaxin-3, interleukin (IL)-8, interferon (IFN)-γ-induced protein-10 (IP-10), monocyte chemoattractant protein (MCP)-1, MCP-4, macrophage-derived chemokines (MDC), macrophage inflammatory protein (MIP)-1α, MIP-1β and thymus- and activation-regulated chemokine (TARC)] or cytokine (IFN-γ, IL-1β, interleukin-2, IL-4, IL-6, IL-10, IL-12p70, IL-13, tumor necrosis factor-α) levels measured were significantly different between the different clinical presentations of CL, as measured by Kruskal-Wallis test. We also compared those with healthy nonendemic controls: our results show a chemokine (IP-10, MCP-1, MCP-4, MDC, MIP-1α, MIP-1β and TARC) but not a cytokine immune signature in patients with CL as compared to healthy nonendemic controls, as measured by Mann-Whitney test. CONCLUSIONS The results of our study did not identify a systemic immune signature or parasite genetic factors associated with different clinical presentation of CL.
Collapse
Affiliation(s)
- Endalew Yizengaw
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Bahir Dar University, Bahir Dar, Ethiopia
- Institute of Biotechnology, Bahir Dar University, Bahir Dar, Ethiopia
- Amhara Public Health Institute, Bahir Dar, Ethiopia
| | - Yegnasew Takele
- Department of Infectious Disease, Imperial College London, London, UK
- Department of Comprehensive Cancer Centre, King's College London, London, UK
| | | | - Bizuayehu Gashaw
- Amhara Public Health Institute, Bahir Dar, Ethiopia
- Department of Biology, College of Science, Bahir Dar University, Bahir Dar, Ethiopia
| | - Mulat Yimer
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Bahir Dar University, Bahir Dar, Ethiopia
- Amhara Public Health Institute, Bahir Dar, Ethiopia
| | | | - Endalkachew Nibret
- Institute of Biotechnology, Bahir Dar University, Bahir Dar, Ethiopia
- Department of Biology, College of Science, Bahir Dar University, Bahir Dar, Ethiopia
| | | | | | - Kefale Ejigu
- Amhara Public Health Institute, Bahir Dar, Ethiopia
| | | | - Abaineh Munshea
- Institute of Biotechnology, Bahir Dar University, Bahir Dar, Ethiopia
- Department of Biology, College of Science, Bahir Dar University, Bahir Dar, Ethiopia
| | - Ingrid Müller
- Department of Infectious Disease, Imperial College London, London, UK
| | - Richard Weller
- Department of Dermatology, University of Edinburgh, Edinburgh, UK
| | - James A Cotton
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
- School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| | - Pascale Kropf
- Department of Infectious Disease, Imperial College London, London, UK.
| |
Collapse
|
6
|
Heggli I, Mengis T, Laux C, Opitz L, Herger N, Menghini D, Schuepbach R, Farshad-Amacker N, Brunner F, Fields A, Farshad M, Distler O, Dudli S. Low back pain patients with Modic type 1 changes exhibit distinct bacterial and non-bacterial subtypes. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100434. [PMID: 38322145 PMCID: PMC10844677 DOI: 10.1016/j.ocarto.2024.100434] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/13/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
Abstract
Objectives Modic type 1 changes (MC1) are vertebral endplate bone marrow (BM) lesions observed on magnetic resonance images in sub-populations of chronic low back pain (CLBP) patients. The etiopathogenesis remains unknown and treatments that modify the underlying pathomechanisms do not exist. We hypothesized that two biological MC1 subtypes exist: a bacterial and a non-bacterial. This would have important implications for developing treatments targeting the underlying pathomechanisms. Methods Intervertebral disc (IVD) samples adjacent to MC1 (n = 34) and control (n = 11) vertebrae were collected from patients undergoing spinal fusion. Cutibacterium acnes (C.acnes) genome copy numbers (GCNs) were quantified in IVD tissues with 16S qPCR, transcriptomic signatures and cytokine profiles were determined in MC1 and control BM by RNA sequencing and immunoassay. Finally, we assessed if C.acnes GCNs are associated with blood plasma cytokines. Results IVD tissues from control levels had <870 C.acnes GCNs/gram IVD. MC1-adjacent IVDs had either "low" (<870) or "high" (>870) C.acnes GCNs. MC1 patients with "high" C.acnes GCNs had upregulated innate immune cell signatures (neutrophil, macrophage/monocyte) and pro-inflammatory cytokines related to neutrophil and macrophage/monocyte function in the BM, consistent with a host defense against bacterium. MC1 patients with "low" C.acnes GCNs had increased adaptive immune cell signatures (T-and B-cell) in the BM and elevated IL-13 blood plasma levels. Conclusion Our study provides the first evidence for the existence of bacterial (C.acnes "high") and non-bacterial (C.acnes "low") subtypes in MC1 patients with CLBP. This supports the need for different treatment strategies.
Collapse
Affiliation(s)
- I. Heggli
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - T. Mengis
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - C.J. Laux
- Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - L. Opitz
- Functional Genomics Center Zurich, University and ETH Zurich, Zurich, Zurich, Switzerland
| | - N. Herger
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - D. Menghini
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - R. Schuepbach
- Unit of Clinical and Applied Research, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - N.A. Farshad-Amacker
- Department of Radiology, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - F. Brunner
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - A.J. Fields
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, USA
| | - M. Farshad
- Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - O. Distler
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| | - S. Dudli
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Physical Medicine and Rheumatology, Balgrist University Hospital, Balgrist Campus, University of Zurich, Zurich, Switzerland
| |
Collapse
|
7
|
Yu L, McGarry S, Cruickshank D, Jensen GS. Rapid increase in immune surveillance and expression of NKT and γδT cell activation markers after consuming a nutraceutical supplement containing Aloe vera gel, extracts of Poria cocos and rosemary. A randomized placebo-controlled cross-over trial. PLoS One 2023; 18:e0291254. [PMID: 37699014 PMCID: PMC10497150 DOI: 10.1371/journal.pone.0291254] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/19/2023] [Indexed: 09/14/2023] Open
Abstract
GOAL To evaluate the acute impact of a nutraceutical blend on immune surveillance. STUDY DESIGN A randomized, double-blind, placebo-controlled, cross-over trial was conducted in 11 healthy subjects. Blood samples were taken immediately before and at 1, 2, and 3 hours after consuming placebo or 500 mg of UP360, which is a blend of botanicals from Aloe vera, Poria cocos, and rosemary (APR extract). Immunophenotyping and flow cytometry quantified numbers of monocytes, NK cells, NKT cells, CD8+ cytotoxic T cells, γδT cells, and total T cells, and expression of CD25 and CD69 activation markers. Plasma was tested for cytokines, chemokines, growth factors, and enzymatic activity of superoxide dismutase and catalase. RESULTS Compared to the placebo, consumption of APR extract triggered rapid increases in chemokine levels starting at 1 hour, including IP-10 (P<0.05) and MCP-1 (P<0.1), which peaked at 2 hours (P<0.01) and 3 hours (P<0.05), respectively. The stem cell-mobilizing growth factor G-CSF increased at 2 hours (P<0.05). Increased immune surveillance involved a transient effect for monocytes at 1 hour, followed by NKT cells, CD8+ cytotoxic T cells, and γδT cells at 2-3 hours. Increased immune cell alertness was seen at 1 hour by increased CD25 expression on monocytes (P<0.01), NKT cells (P<0.01), and T cells (P<0.05). NKT cells showed upregulation of CD69 at 2 hours (P<0.01). Increased enzymatic activity was seen at 2 hours for the antioxidant enzymes superoxide dismutase (P<0.05) and catalase (P<0.01). CONCLUSION Consumption of APR extract triggered acute changes to chemokine levels. In addition, immune alertness was increased via the expression of activation markers on multiple types of innate immune cells, followed by increased immune surveillance and antioxidant protection. This suggests a beneficial enhancement of natural immune surveillance, likely via a combination of gut-mediated cytokine release and vagus nerve communication, in combination with cellular protection from oxidative stress.
Collapse
Affiliation(s)
- Liu Yu
- NIS Labs, Port Dover, Ontario, Canada
| | | | | | | |
Collapse
|
8
|
Oliveira D, Lopes KF, de Avelar DM, Cota G, Oliveira E. Accuracy of serological tests in diagnosing mucosal leishmaniasis. Parasitol Res 2023; 122:2001-2010. [PMID: 37391643 DOI: 10.1007/s00436-023-07900-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/12/2023] [Indexed: 07/02/2023]
Abstract
In this serum panel-based study, we evaluated the accuracy of serological tests originally developed for visceral leishmaniasis (VL), for diagnosis of mucosal leishmaniasis (ML). A total of five tests were evaluated, four of which are registered at the National Agency of Sanitary Surveillance (Agência Nacional de Vigilância Sanitária-ANVISA) (RIDASCREEN® Leishmania Ab from R-Biopharm AG., Leishmania ELISA IgG + IgM from Vircell S.L., IFI Leishmaniose Humana-BioManguinhos, and IT-LEISH® from Bio-Rad Laboratories, Inc.), and the other a direct agglutination test (DAT-LPC) prototype kit developed at Fiocruz. The panel was composed of 40 serum samples from patients with confirmed ML and 20 from patients with mucosal involvement and negative parasitological/molecular tests for leishmaniasis and confirmation of another etiology. All cases were treated from 2009 to 2016 in a referral center for leishmaniasis in Belo Horizonte, Minas Gerais, Brazil (Instituto René Rachou, Fiocruz). Diagnostic accuracy, based on the cut-off point for VL diagnosis, was 86.2% with RIDASCREEN® Leishmania Ab, 73.3% with Leishmania ELISA IgG + IgM, and 66.7% with IFI Leishmaniose Humana, while IT-LEISH® and DAT-LPC had the lowest accuracy (38.3%), despite high specificity (100% and 95%, respectively). New cut-off points defined with sera from ML patients improved accuracy from 86.2 to 89% (p = 0.64) and 73.3 to 88% (p = 0.04) for RIDASCREEN® Leishmania Ab and Leishmania ELISA IgG + IgM, respectively. Moreover, these tests presented greater sensitivity and immunoreactivity in patients with moderate/severe clinical ML forms. The data of this study suggest that ELISA assays can contribute to laboratory diagnosis, especially for patients with moderate or severe mucosal involvement.
Collapse
Affiliation(s)
- Diana Oliveira
- Clinical Research and Public Politics in Infectious and Parasitic Diseases, Instituto René Rachou, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Karine Ferreira Lopes
- Clinical Research and Public Politics in Infectious and Parasitic Diseases, Instituto René Rachou, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Daniel Moreira de Avelar
- Clinical Research and Public Politics in Infectious and Parasitic Diseases, Instituto René Rachou, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Gláucia Cota
- Clinical Research and Public Politics in Infectious and Parasitic Diseases, Instituto René Rachou, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil.
| | - Edward Oliveira
- Clinical Research and Public Politics in Infectious and Parasitic Diseases, Instituto René Rachou, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
9
|
Bruschi F, Pinto B, Fallahi P, Ferrari SM, Antonelli A. Increased neutrophil derived chemokines (CXCL10 and CCL2) in human trichinellosis as possible serological markers of the polarization of the immune response against the parasite. Cytokine 2023; 166:156205. [PMID: 37058963 DOI: 10.1016/j.cyto.2023.156205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 04/16/2023]
Abstract
Trichinella britovi is a widely distributed parasitic nematode, transmitted through ingestion of raw or poorly cooked meat containing muscle larvae. This helminth can regulate the host immune system during the early phase of infection. The immune mechanism mainly involves the interaction of Th1 and Th2 responses and related cytokines. Chemokines (C-X-C or C-C) and matrix metalloproteinases (MMPs) have also shown to be implicated in a number of parasitic infections, mainly malaria, neurocysticercosis, angiostronyloidosis, and schistosomiasis, but poor is known about their role in human Trichinella infection. We previously found that serum MMP-9 levels were significantly increased in T. britovi infected patients with relevant symptoms such as diarrhea, myalgia, and facial oedema, which makes these enzymes a potential reliable indicator of inflammation in trichinellosis patients. These changes were also observed in T. spiralis/T. pseudospiralis experimentally infected mice. No data are available about circulating levels of two pro-inflammatory chemokines, CXCL10 and CCL2, in trichinellosis patients with or w/o clinical signs of the infection. In this study, the association of serum level of CXCL10 and CCL2 with clinical outcome of T. britovi infection and their relation to MMP-9 were investigated. Patients (median age 49 ± 0.33 years) acquired infection by consuming raw sausages prepared with wild boar and pork meat. Sera were collected during the acute and the convalescent phases of the infection. A positive significant association (r = 0.61, p = 0.0004) was observed between MMP-9 and CXCL10 levels. The CXCL10 level significantly correlated with the severity of symptoms in patients being particularly higher in patients suffering diarrhea, myalgia, and facial oedema, thus suggesting a positive association of this chemokine with symptomatologic traits, especially myalgia (and increased LDH and CPK levels) (p < 0.005). No correlation was found between levels of CCL2 and the clinical symptoms.
Collapse
Affiliation(s)
- Fabrizio Bruschi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy.
| | - Barbara Pinto
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Poupak Fallahi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | | | - Alessandro Antonelli
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
10
|
Kumar R, Bhatia M, Pai K. Role of Chemokines in the Pathogenesis of Visceral Leishmaniasis. Curr Med Chem 2022; 29:5441-5461. [PMID: 35579167 DOI: 10.2174/0929867329666220509171244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 12/23/2021] [Accepted: 03/02/2022] [Indexed: 11/22/2022]
Abstract
Visceral leishmaniasis (VL; also known as kala-azar), caused by the protozoan parasite Leishmania donovani, is characterized by the inability of the host to generate an effective immune response. The manifestations of the disease depend on the involvement of various immune components such as activation of macrophages, cell mediated immunity, secretion of cytokines and chemokines, etc. Macrophages are the final host cells for Leishmania parasites to multiply, and they are the key to a controlled or aggravated response that leads to clinical symptoms. The two most common macrophage phenotypes are M1 and M2. The pro-inflammatory microenvironment (mainly by IL-1β, IL-6, IL-12, IL-23, and TNF-α cytokines) and tissue injury driven by classically activated macrophages (M1-like) and wound healing driven by alternatively activated macrophages (M2-like) in an anti-inflammatory environment (mainly by IL-10, TGF-β, chemokine ligand (CCL)1, CCL2, CCL17, CCL18, and CCL22). Moreover, on polarized Th cells, chemokine receptors are expressed differently. Typically, CXCR3 and CCR5 are preferentially expressed on polarized Th1 cells, whereas CCR3, CCR4, and CCR8 have been associated with the Th2 phenotype. Further, the ability of the host to produce a cell-mediated immune response capable of regulating and/or eliminating the parasite is critical in the fight against the disease. Here, we review the interactions between parasites and chemokines and chemokine receptors in the pathogenesis of VL.
Collapse
Affiliation(s)
- Ramesh Kumar
- Department of Biochemistry, Bundelkhand University, Jhansi (UP), India
| | - Madhav Bhatia
- Department of Pathology and Biomedical Sciences, University of Otago, Christchurch, New Zealand
| | - Kalpana Pai
- Department of Zoology, Savitribai Phule Pune University, Pune, Maharashtra, India
| |
Collapse
|
11
|
Chemokines in Leishmaniasis: Map of cell movements highlights the landscape of infection and pathogenesis. Cytokine 2021; 147:155339. [DOI: 10.1016/j.cyto.2020.155339] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
|
12
|
Carneiro MB, Peters NC. The Paradox of a Phagosomal Lifestyle: How Innate Host Cell- Leishmania amazonensis Interactions Lead to a Progressive Chronic Disease. Front Immunol 2021; 12:728848. [PMID: 34557194 PMCID: PMC8452962 DOI: 10.3389/fimmu.2021.728848] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Intracellular phagosomal pathogens represent a formidable challenge for innate immune cells, as, paradoxically, these phagocytic cells can act as both host cells that support pathogen replication and, when properly activated, are the critical cells that mediate pathogen elimination. Infection by parasites of the Leishmania genus provides an excellent model organism to investigate this complex host-pathogen interaction. In this review we focus on the dynamics of Leishmania amazonensis infection and the host innate immune response, including the impact of the adaptive immune response on phagocytic host cell recruitment and activation. L. amazonensis infection represents an important public health problem in South America where, distinct from other Leishmania parasites, it has been associated with all three clinical forms of leishmaniasis in humans: cutaneous, muco-cutaneous and visceral. Experimental observations demonstrate that most experimental mouse strains are susceptible to L. amazonensis infection, including the C57BL/6 mouse, which is resistant to other species such as Leishmania major, Leishmania braziliensis and Leishmania infantum. In general, the CD4+ T helper (Th)1/Th2 paradigm does not sufficiently explain the progressive chronic disease established by L. amazonensis, as strong cell-mediated Th1 immunity, or a lack of Th2 immunity, does not provide protection as would be predicted. Recent findings in which the balance between Th1/Th2 immunity was found to influence permissive host cell availability via recruitment of inflammatory monocytes has also added to the complexity of the Th1/Th2 paradigm. In this review we discuss the roles played by innate cells starting from parasite recognition through to priming of the adaptive immune response. We highlight the relative importance of neutrophils, monocytes, dendritic cells and resident macrophages for the establishment and progressive nature of disease following L. amazonensis infection.
Collapse
Affiliation(s)
- Matheus B Carneiro
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Nathan C Peters
- Snyder Institute for Chronic Diseases, Departments of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine and Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
13
|
Niu F, Liao K, Hu G, Moidunny S, Roy S, Buch S. HIV Tat-Mediated Induction of Monocyte Transmigration Across the Blood-Brain Barrier: Role of Chemokine Receptor CXCR3. Front Cell Dev Biol 2021; 9:724970. [PMID: 34527676 PMCID: PMC8435688 DOI: 10.3389/fcell.2021.724970] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/10/2021] [Indexed: 01/17/2023] Open
Abstract
HIV trans-activator of transcription (Tat), one of the cytotoxic proteins secreted from HIV-infected cells, is also known to facilitate chemokine-mediated transmigration of monocytes into the brain leading, in turn, to neuroinflammation and thereby contributing to the development of HIV-associated neurocognitive disorders (HAND). The mechanism(s) underlying HIV Tat-mediated enhancement of monocyte transmigration, however, remain largely unknown. CXC chemokine receptor 3 (CXCR3) that is expressed by the peripheral monocytes is known to play a role in the monocyte influx and accumulation. In the present study, we demonstrate for the first time that exposure of human monocytes to HIV Tat protein resulted in upregulated expression of CXCR3 leading, in turn, to increased monocyte transmigration across the blood–brain barrier (BBB) both in the in vitro and in vivo model systems. This process involved activation of toll-like receptor 4 (TLR4), with downstream phosphorylation and activation of TANK-binding kinase 1 (TBK1), and subsequent phosphorylation and nuclear translocation of interferon regulatory factor 3 (IRF3), ultimately leading to enhanced expression of CXCR3 in human monocytes. These findings imply a novel molecular mechanism underlying HIV Tat-mediated increase of monocyte transmigration across the BBB, while also implicating a novel role of CXCR3-dependent monocyte transmigration in HIV Tat-mediated neuroinflammation.
Collapse
Affiliation(s)
- Fang Niu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States.,Division of Clinical Research and Evaluative Sciences, Department of Medicine, Creighton University, Omaha, NE, United States
| | - Ke Liao
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shamsudheen Moidunny
- Department of Surgery, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Sabita Roy
- Department of Surgery, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
14
|
Zheng WB, Zou Y, Liu Q, Hu MH, Elsheikha HM, Zhu XQ. Toxocara canis Infection Alters lncRNA and mRNA Expression Profiles of Dog Bone Marrow. Front Cell Dev Biol 2021; 9:688128. [PMID: 34277631 PMCID: PMC8277978 DOI: 10.3389/fcell.2021.688128] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/31/2021] [Indexed: 01/05/2023] Open
Abstract
Bone marrow is the main hematopoietic organ that produces red blood cells, granulocytes, monocyte/macrophages, megakaryocytes, lymphocytes, and myeloid dendritic cells. Many of these cells play roles in the pathogenesis of Toxocara canis infection, and understanding how infection alters the dynamics of transcription regulation in bone marrow is therefore critical for deciphering the global changes in the dog transcriptional signatures during T. canis infection. In this study, long non-coding RNA (lncRNA) and messenger RNA (mRNA) expression profiles in the bone marrow of Beagle dogs infected with T. canis were determined at 12 h post-infection (hpi), 24 hpi, 96 hpi, and 36 days post-infection (dpi). RNA-sequencing and bioinformatics analysis identified 1,098, 984, 1,120, and 1,305 differentially expressed lncRNAs (DElncRNAs), and 196, 253, 223, and 328 differentially expressed mRNAs (DEmRNAs) at 12 h, 24 h, 96 h, and 36 days after infection, respectively. We also identified 29, 36, 38, and 68 DEmRNAs potentially cis-regulated by 44, 44, 51, and 80 DElncRNAs at 12 hpi, 24 hpi, 96 hpi, and 36 dpi, respectively. To validate the sequencing findings, qRT-PCR was performed on 10 randomly selected transcripts. Many altered genes were involved in the differentiation of bone marrow cells. GO of DElncRNAs and GO and KEGG pathway analyses of DEmRNAs revealed alterations in several signaling pathways, including pathways involved in energy metabolism, amino acid biosynthesis and metabolism, Wnt signaling pathway, Huntington's disease, HIF-1 signaling pathway, cGMP–PKG signaling pathway, dilated cardiomyopathy, and adrenergic signaling in cardiomyocytes. These findings revealed that bone marrow of T. canis-infected dogs exhibits distinct lncRNA and mRNA expression patterns compared to healthy control dogs. Our data provide novel insights into T. canis interaction with the definitive host and shed light on the significance of the non-coding portion of the dog genome in the pathogenesis of toxocariasis.
Collapse
Affiliation(s)
- Wen-Bin Zheng
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yang Zou
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Qing Liu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Min-Hua Hu
- National Canine Laboratory Animal Resource Center, Guangzhou General Pharmaceutical Research Institute Co., Ltd, Guangzhou, China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Loughborough, United Kingdom
| | - Xing-Quan Zhu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China.,State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Key Laboratory of Veterinary Public Health of Higher Education of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
15
|
Dutra BM, Rodrigues NLDC, Fonseca FRM, de Moura TR, Pacheco de Almeida R, de Jesus AR, Abreu TM, Pompeu MMDL, Teixeira CR, Teixeira MJ. CXCL10 immunomodulatory effect against infection caused by an antimony refractory isolate of Leishmania braziliensis in mice. Parasite Immunol 2020; 43:e12805. [PMID: 33131089 DOI: 10.1111/pim.12805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 10/16/2020] [Accepted: 10/19/2020] [Indexed: 11/28/2022]
Abstract
Leishmania braziliensis is the main causative agent of American tegumentary leishmaniasis in Brazil. Current treatment includes different drugs that have important side effects and identification of cases of parasite resistance to treatment support the search for new therapeutic strategies. Recent findings have indicated that CXCL10, a chemokine that recruits and activates Th1 cells, NK cells, macrophages, dendritic cells and B lymphocytes, is a potential alternative to treat Leishmania infection. Here, we tested CXCL10 immunotherapy against experimental infection caused by an antimony-resistant isolate of Leishmania braziliensis. Following infection, mice were treated with CXCL10 for 7 days after onset of lesions. We demonstrate that mice treated with CXCL10 controlled lesion progression and parasite burden more efficiently comparing to controls. An increased IFN-γ, IL-10, TGF-β and low IL-4 production combined with a distinct inflammatory infiltrate composed by activated macrophages, lymphocytes and granulomas was observed in the CXCL10-treated group comparing to controls. However, CXCL10 and Glucantime combined therapy did not improve CXCL10-induced protective effect. Our findings reinforce the potential of CXCL10 immunotherapy as an alternative treatment against infection caused by L. braziliensis resistant to conventional chemotherapy.
Collapse
Affiliation(s)
- Brunheld Maia Dutra
- Department of Pathology and Legal Medicine, Federal University of Ceará, Fortaleza, Brazil
| | | | | | | | | | - Amélia Ribeiro de Jesus
- Department of Internal Medicine and Pathology, Federal University of Sergipe, Aracajú, Brazil
| | - Ticiana Monteiro Abreu
- Department of Pathology and Legal Medicine, Federal University of Ceará, Fortaleza, Brazil
| | | | | | - Maria Jania Teixeira
- Department of Pathology and Legal Medicine, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
16
|
Cardoso FDO, Zaverucha-do-Valle T, Almeida-Souza F, Abreu-Silva AL, Calabrese KDS. Modulation of Cytokines and Extracellular Matrix Proteins Expression by Leishmania amazonensis in Susceptible and Resistant Mice. Front Microbiol 2020; 11:1986. [PMID: 32983013 PMCID: PMC7487551 DOI: 10.3389/fmicb.2020.01986] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/27/2020] [Indexed: 12/24/2022] Open
Abstract
Leishmaniases are a complex of diseases with a broad spectrum of clinical forms, which depend on the parasite species, immunological status, and genetic background of the host. In the Leishmania major model, susceptibility is associated with the Th2 pattern of cytokines production, while resistance is associated with Th1 response. However, the same dichotomy does not occur in L. amazonensis-infected mice. Cytokines are key players in these diseases progression, while the extracellular matrix (ECM) components participate in the process of parasite invasion as well as lesion healing. In this article, we analyzed the influence of host genetics on the expression of cytokines, inducible nitric oxide synthase (iNOS), and ECM proteins, as well as the parasite load in mice with different genetic backgrounds infected by L. amazonensis. C57BL/10 and C3H/He mice were subcutaneously infected with 106L. amazonensis promastigotes. Lesion kinetics, parasite load, cytokines, iNOS, and ECM proteins expression were measured by quantitative PCR (qPCR) in the footpad, draining lymph nodes, liver, and spleen at early (24 h and 30 days) and late phase (120 and 180 days) of infection. Analysis of lesion kinetics showed that C57BL/10 mice developed ulcerative lesions at the inoculation site after L. amazonensis infection, while C3H/He showed slight swelling in the footpad 180 days after infection. C57BL/10 showed progressive enhancement of parasite load in all analyzed organs, while C3H/He mice showed extremely low parasite loads. Susceptible C57BL/10 mice showed high levels of TGF-β mRNA in the footpad early in infection and high levels of proinflammatory cytokines mRNA (IL-12, TNF-α, and IFN-γ) and iNOS in the late phase of the infection. There is an association between increased expression of fibronectin, laminin, collagen III and IV, and TGF-β. On the other hand, resistant C3H/He mice presented a lower repertory of cytokines mRNA expression when compared with susceptible C57BL/10 mice, basically producing TNF-α, collagen IV, and laminin early in infection. The findings of our study indicate that L. amazonensis infection induces different cytokine expression in resistant and susceptible mice but not like the L. major model. An organ-compartmentalized cytokine response was observed in our model. Host genetics determine this response, which modulates ECM proteins expression.
Collapse
Affiliation(s)
- Flávia de Oliveira Cardoso
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Tânia Zaverucha-do-Valle
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Fernando Almeida-Souza
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.,Laboratório de Anatomopatologia, Departamento de Patologia, Universidade Estadual do Maranhão, São Luís, Brazil
| | - Ana Lúcia Abreu-Silva
- Laboratório de Anatomopatologia, Departamento de Patologia, Universidade Estadual do Maranhão, São Luís, Brazil
| | - Kátia da Silva Calabrese
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
17
|
Evaluation of the Ability of Miltefosine Associated with Topical GM-CSF in Modulating the Immune Response of Patients with Cutaneous Leishmaniasis. J Immunol Res 2020; 2020:2789859. [PMID: 32851099 PMCID: PMC7439779 DOI: 10.1155/2020/2789859] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 07/09/2020] [Accepted: 07/17/2020] [Indexed: 11/17/2022] Open
Abstract
Cutaneous leishmaniasis (CL) due to L. braziliensis is associated with an exaggerated inflammatory response and tissue damage. Miltefosine is more effective than pentavalent antimony (Sbv) in the treatment of CL, and here, we evaluate the ability of Sbv, miltefosine, and GM-CSF administered intravenously, orally, or topically, respectively, to modify the immune response. Patients were treated with miltefosine plus GM-CSF, miltefosine plus placebo, or Sbv. Mononuclear cells were stimulated with soluble Leishmania antigen (SLA) on day 0 and day 15 of therapy, and cytokine levels were determined in supernatants by ELISA. The lymphocyte proliferation and oxidative burst were evaluated by flow cytometry, and the degree of infection and Leishmania killing by optical microscopy. Proliferation of CD4+ T cells were enhanced in patients using miltefosine and in CD8+ T cells when GM-CSF was associated. Enhancement in the oxidative burst occurred in the miltefosine plus GM-CSF group on day 15 of therapy. Moreover, the number of L. braziliensis in infected monocytes on day 15 as well as the percentage of infected cells was lower after 48- and 72-hour culture in cells from patients treated with miltefosine plus GM-CSF. In addition to the ability of miltefosine to kill Leishmania, the changes in the immune response caused by miltefosine and GM-CSF may increase the cure rate of CL patients using these drugs.
Collapse
|
18
|
The role of monocytes/macrophages in Leishmania infection: A glance at the human response. Acta Trop 2020; 207:105456. [PMID: 32222362 DOI: 10.1016/j.actatropica.2020.105456] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 12/18/2022]
Abstract
Leishmania are obligate intracellular parasites of mononuclear phagocytes transmitted by Phlebotomine sandflies. Monocytes are one of the main cell types recruited to the site of the bite having an important role in the defense against Leishmania parasites in the first hours of infection. In the tissue, macrophages play a pivotal role as both the primary replication sites and the major effector cells responsible for parasite elimination. Many authors have reviewed the monocyte/macrophage-Leishmania interactions from results derived in mice, however, given the important differences between mice an humans we considered vital to discuss the role of these cells in human leishmaniasis. In this review, we recapitulated the most important studies carried out to understand the different roles of human monocyte/macrophages in Leishmania infection and how they can participate in both control and the immunopathogenesis of the disease.
Collapse
|
19
|
Carneiro MB, Lopes ME, Hohman LS, Romano A, David BA, Kratofil R, Kubes P, Workentine ML, Campos AC, Vieira LQ, Peters NC. Th1-Th2 Cross-Regulation Controls Early Leishmania Infection in the Skin by Modulating the Size of the Permissive Monocytic Host Cell Reservoir. Cell Host Microbe 2020; 27:752-768.e7. [PMID: 32298657 DOI: 10.1016/j.chom.2020.03.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 01/13/2020] [Accepted: 03/16/2020] [Indexed: 12/19/2022]
Abstract
The impact of T helper (Th) 1 versus Th2 immunity on intracellular infections is attributed to classical versus alternative activation of macrophages leading to resistance or susceptibility. However, observations in multiple infectious settings demonstrate deficiencies in mediators of Th1-Th2 immunity, which have paradoxical or no impact. We report that prior to influencing activation, Th1/Th2 immunity first controls the size of the permissive host cell reservoir. During early Leishmania infection of the skin, IFN-γ- or STAT6-mediated changes in phagocyte activation were counteracted by changes in IFN-γ-mediated recruitment of permissive CCR2+ monocytes. Monocytes were required for early parasite expansion and acquired an alternatively activated phenotype despite the Th1 dermal environment required for their recruitment. Surprisingly, STAT6 did not enhance intracellular parasite proliferation, but rather modulated the size and permissiveness of the monocytic host cell reservoir via regulation of IFN-γ and IL-10. These observations expand our understanding of the Th1-Th2 paradigm during infection.
Collapse
Affiliation(s)
- Matheus Batista Carneiro
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6, Canada; Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Mateus Eustáquio Lopes
- Departamento de Bioquímica e Imunologia - ICB - Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270901, Brazil
| | - Leah S Hohman
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6, Canada; Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Audrey Romano
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bruna Araujo David
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6, Canada; Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Rachel Kratofil
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6, Canada; Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Paul Kubes
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6, Canada; Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Matthew L Workentine
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Alexandre C Campos
- Departamento de Bioquímica e Imunologia - ICB - Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270901, Brazil
| | - Leda Quercia Vieira
- Departamento de Bioquímica e Imunologia - ICB - Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270901, Brazil
| | - Nathan C Peters
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6, Canada; Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada.
| |
Collapse
|
20
|
Kazer SW, Aicher TP, Muema DM, Carroll SL, Ordovas-Montanes J, Miao VN, Tu AA, Ziegler CGK, Nyquist SK, Wong EB, Ismail N, Dong M, Moodley A, Berger B, Love JC, Dong KL, Leslie A, Ndhlovu ZM, Ndung'u T, Walker BD, Shalek AK. Integrated single-cell analysis of multicellular immune dynamics during hyperacute HIV-1 infection. Nat Med 2020; 26:511-518. [PMID: 32251406 PMCID: PMC7237067 DOI: 10.1038/s41591-020-0799-2] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 02/12/2020] [Indexed: 02/07/2023]
Abstract
Cellular immunity is critical for controlling intracellular pathogens, but individual cellular dynamics and cell-cell cooperativity in evolving human immune responses remain poorly understood. Single-cell RNA-sequencing (scRNA-seq) represents a powerful tool for dissecting complex multicellular behaviors in health and disease1,2 and nominating testable therapeutic targets3. Its application to longitudinal samples could afford an opportunity to uncover cellular factors associated with the evolution of disease progression without potentially confounding inter-individual variability4. Here, we present an experimental and computational methodology that uses scRNA-seq to characterize dynamic cellular programs and their molecular drivers, and apply it to HIV infection. By performing scRNA-seq on peripheral blood mononuclear cells from four untreated individuals before and longitudinally during acute infection5, we were powered within each to discover gene response modules that vary by time and cell subset. Beyond previously unappreciated individual- and cell-type-specific interferon-stimulated gene upregulation, we describe temporally aligned gene expression responses obscured in bulk analyses, including those involved in proinflammatory T cell differentiation, prolonged monocyte major histocompatibility complex II upregulation and persistent natural killer (NK) cell cytolytic killing. We further identify response features arising in the first weeks of infection, for example proliferating natural killer cells, which potentially may associate with future viral control. Overall, our approach provides a unified framework for characterizing multiple dynamic cellular responses and their coordination.
Collapse
Affiliation(s)
- Samuel W Kazer
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Toby P Aicher
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel M Muema
- African Health Research Institute, Durban, South Africa
- HIV Pathogenesis Programme, Nelson R. Mandela School of Medicine, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Shaina L Carroll
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Jose Ordovas-Montanes
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Vincent N Miao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School & Massachusetts Institute of Technology, Boston, MA, USA
| | - Ang A Tu
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Carly G K Ziegler
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School & Massachusetts Institute of Technology, Boston, MA, USA
| | - Sarah K Nyquist
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emily B Wong
- African Health Research Institute, Durban, South Africa
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
- Division of Infection and Immunity, University College London, London, UK
- Harvard Medical School, Boston, MA, USA
| | - Nasreen Ismail
- HIV Pathogenesis Programme, Nelson R. Mandela School of Medicine, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Mary Dong
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Amber Moodley
- HIV Pathogenesis Programme, Nelson R. Mandela School of Medicine, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - J Christopher Love
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Krista L Dong
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Alasdair Leslie
- African Health Research Institute, Durban, South Africa
- Division of Infection and Immunity, University College London, London, UK
| | - Zaza M Ndhlovu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- African Health Research Institute, Durban, South Africa
- HIV Pathogenesis Programme, Nelson R. Mandela School of Medicine, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Thumbi Ndung'u
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- African Health Research Institute, Durban, South Africa
- HIV Pathogenesis Programme, Nelson R. Mandela School of Medicine, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
- Division of Infection and Immunity, University College London, London, UK
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Bruce D Walker
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- HIV Pathogenesis Programme, Nelson R. Mandela School of Medicine, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | - Alex K Shalek
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Program in Health Sciences and Technology, Harvard Medical School & Massachusetts Institute of Technology, Boston, MA, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Program in Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
21
|
O D, Waelkens E, Vanhie A, Peterse D, Fassbender A, D'Hooghe T. The Use of Antibody Arrays in the Discovery of New Plasma Biomarkers for Endometriosis. Reprod Sci 2020; 27:751-762. [PMID: 32016799 DOI: 10.1007/s43032-019-00081-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/05/2019] [Indexed: 12/21/2022]
Abstract
A noninvasive diagnostic test for endometriosis is needed to shorten the current diagnostic delay of 8-11 years. The goal of this study was to discover new biomarkers for endometriosis using an antibody array approach. A total of 103 plasma samples from patients with laparoscopically confirmed presence (n = 68) or absence (n = 35) of endometriosis were selected. Samples were pooled according to disease status, cycle phase, disease stage, and phenotype. Pooled samples were screened for possible biomarkers using the L-series 1000 and Quantibody 660 arrays from RayBiotech. Technical verification of ten markers was done using a custom-made multiplex immunoassay identifying ten proteins (10-plex) and later by single ELISA. Due to the limited reproducibility of the L-series 1000 immunoassay, the biomarker screening was performed using the Quantibody 660, a sandwich-based multiplex immunoassay, which showed that 280 proteins were upregulated, and 29 proteins downregulated in the endometriosis pool versus the control pool. In order to assess the reproducibility of these results, ten preselected proteins were analyzed using a custom 10-plex. Four proteins (CD48, DNAM-1, IL-31, and XIAP) were confirmed to be differentially expressed when comparing the endometriosis and control pool. However, only IL-31 showed a univariate statistical difference between endometriosis and control groups in individual samples that were part of the initial pools. In conclusion, discovery and verification of potential markers proved challenging using multiplex immunoassay methods, mainly due to issues with reproducibility. Only IL-31 showed potential as possible biomarker for endometriosis.
Collapse
Affiliation(s)
- Dorien O
- Department of Development and Regeneration, Woman and Child, KU Leuven, Leuven, Belgium
| | - Etienne Waelkens
- Facility for systems biology based mass spectrometry, KU Leuven, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Arne Vanhie
- Department of Development and Regeneration, Woman and Child, KU Leuven, Leuven, Belgium
| | - Daniëlle Peterse
- Department of Development and Regeneration, Woman and Child, KU Leuven, Leuven, Belgium
| | - Amelie Fassbender
- Department of Development and Regeneration, Woman and Child, KU Leuven, Leuven, Belgium
| | - Thomas D'Hooghe
- Department of Development and Regeneration, Woman and Child, KU Leuven, Leuven, Belgium.
| |
Collapse
|
22
|
Rodrigues V, André S, Maksouri H, Mouttaki T, Chiheb S, Riyad M, Akarid K, Estaquier J. Transcriptional Analysis of Human Skin Lesions Identifies Tryptophan-2,3-Deoxygenase as a Restriction Factor for Cutaneous Leishmania. Front Cell Infect Microbiol 2019; 9:338. [PMID: 31637219 PMCID: PMC6788307 DOI: 10.3389/fcimb.2019.00338] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 09/18/2019] [Indexed: 12/18/2022] Open
Abstract
Disease manifestation after infection with cutaneous Leishmania species is the result of a complex interplay of diverse factors, including the immune status of the host, the infecting parasite species, or the parasite load at the lesion site. Understanding how these factors impact on the pathology of cutaneous leishmaniasis (CL) may provide new targets to manage the infection and improve clinical outcome. We quantified the relative expression of 170 genes involved in a diverse range of biological processes, in the skin biopsies from patients afflicted with CL caused by infection with either L. major or L. tropica. As compared to healthy skin, CL lesions bear elevated levels of transcripts involved in the immune response, and conversely, present a significant downregulation in the expression of genes involved in epidermal integrity and arginine or fatty acid metabolism. The expression of transcripts encoding for cytotoxic mediators and chemokines in lesions was inversely correlated with the expression of genes involved in epidermal integrity, suggesting that cytotoxicity is a major mediator of CL pathology. When comparing the transcriptional profiles of lesions caused by either L. major or L. tropica, we found them to be very similar, the later presenting an aggravated inflammatory/cytotoxic profile. Finally, we identified genes positively correlated with the parasite load in lesions. Among others, these included Th2 or regulatory cytokines, such as IL4 or IL10. Remarkably, a single gene among our dataset, encoding for tryptophan-2,3-deoxygenase (TDO), presented a negative correlation with the parasite load, suggesting that its expression may restrict parasite numbers in lesions. In agreement, treatment of macrophages infected with L. major in vitro with a TDO inhibitor led to an increase in parasite transcripts. Our work provides new insights into the factors that impact CL pathology and identifies TDO as a restriction factor for cutaneous Leishmania.
Collapse
Affiliation(s)
| | - Sónia André
- CNRS-ERL3649, Université Paris Descartes, Paris, France
| | - Hasnaa Maksouri
- Research Team on Immunopathology of Infectious and Systemic Diseases, Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca, Morocco
| | - Tarik Mouttaki
- Research Team on Immunopathology of Infectious and Systemic Diseases, Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca, Morocco
| | - Soumiya Chiheb
- Research Team on Immunopathology of Infectious and Systemic Diseases, Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca, Morocco.,Department of Dermatology, University Hospital Ibn Rochd of Casablanca, Casablanca, Morocco
| | - Myriam Riyad
- Research Team on Immunopathology of Infectious and Systemic Diseases, Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca, Morocco.,Laboratory of Parasitology, Faculty of Medicine and Pharmacy, University of Hassan II Casablanca (UH2C), Casablanca, Morocco
| | - Khadija Akarid
- Molecular Genetics and Immunophysiopathology Research Team, Health and Environment Laboratory, Aïn Chock Faculty of Sciences, University of Hassan II Casablanca (UH2C), Casablanca, Morocco
| | - Jérôme Estaquier
- CNRS-ERL3649, Université Paris Descartes, Paris, France.,Faculty of Medicine, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Laval University, Quebec, QC, Canada
| |
Collapse
|
23
|
Antonia AL, Gibbs KD, Trahair ED, Pittman KJ, Martin AT, Schott BH, Smith JS, Rajagopal S, Thompson JW, Reinhardt RL, Ko DC. Pathogen Evasion of Chemokine Response Through Suppression of CXCL10. Front Cell Infect Microbiol 2019; 9:280. [PMID: 31440475 PMCID: PMC6693555 DOI: 10.3389/fcimb.2019.00280] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/23/2019] [Indexed: 01/10/2023] Open
Abstract
Clearance of intracellular pathogens, such as Leishmania (L.) major, depends on an immune response with well-regulated cytokine signaling. Here we describe a pathogen-mediated mechanism of evading CXCL10, a chemokine with diverse antimicrobial functions, including T cell recruitment. Infection with L. major in a human monocyte cell line induced robust CXCL10 transcription without increasing extracellular CXCL10 protein concentrations. We found that this transcriptionally independent suppression of CXCL10 is mediated by the virulence factor and protease, glycoprotein-63 (gp63). Specifically, GP63 cleaves CXCL10 after amino acid A81 at the base of a C-terminal alpha-helix. Cytokine cleavage by GP63 demonstrated specificity, as GP63 cleaved CXCL10 and its homologs, which all bind the CXCR3 receptor, but not distantly related chemokines, such as CXCL8 and CCL22. Further characterization demonstrated that CXCL10 cleavage activity by GP63 was produced by both extracellular promastigotes and intracellular amastigotes. Crucially, CXCL10 cleavage impaired T cell chemotaxis in vitro, indicating that cleaved CXCL10 cannot signal through CXCR3. Ultimately, we propose CXCL10 suppression is a convergent mechanism of immune evasion, as Salmonella enterica and Chlamydia trachomatis also suppress CXCL10. This commonality suggests that counteracting CXCL10 suppression may provide a generalizable therapeutic strategy against intracellular pathogens. Importance Leishmaniasis, an infectious disease that annually affects over one million people, is caused by intracellular parasites that have evolved to evade the host's attempts to eliminate the parasite. Cutaneous leishmaniasis results in disfiguring skin lesions if the host immune system does not appropriately respond to infection. A family of molecules called chemokines coordinate recruitment of the immune cells required to eliminate infection. Here, we demonstrate a novel mechanism that Leishmania (L.) spp. employ to suppress host chemokines: a Leishmania-encoded protease cleaves chemokines known to recruit T cells that fight off infection. We observe that other common human intracellular pathogens, including Chlamydia trachomatis and Salmonella enterica, reduce levels of the same chemokines, suggesting a strong selective pressure to avoid this component of the immune response. Our study provides new insights into how intracellular pathogens interact with the host immune response to enhance pathogen survival.
Collapse
Affiliation(s)
- Alejandro L. Antonia
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, United States
| | - Kyle D. Gibbs
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, United States
| | - Esme D. Trahair
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, United States
| | - Kelly J. Pittman
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, United States
| | - Amelia T. Martin
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, United States
| | - Benjamin H. Schott
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, United States
| | - Jeffrey S. Smith
- Department of Biochemistry, School of Medicine, Duke University, Durham, NC, United States
| | - Sudarshan Rajagopal
- Department of Biochemistry, School of Medicine, Duke University, Durham, NC, United States
- Division of Cardiology, Department of Medicine, School of Medicine, Duke University, Durham, NC, United States
| | - J. Will Thompson
- Proteomics and Metabolomics Shared Resource, Center for Genomics and Computational Biology, School of Medicine, Duke University, Durham, NC, United States
| | - Richard Lee Reinhardt
- Department of Biomedical Research, National Jewish Health, Denver, CO, United States
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Dennis C. Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, United States
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Duke University, Durham, NC, United States
| |
Collapse
|
24
|
MicroRNA 155 Contributes to Host Immunity against Leishmania donovani but Is Not Essential for Resolution of Infection. Infect Immun 2019; 87:IAI.00307-19. [PMID: 31182615 DOI: 10.1128/iai.00307-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 05/27/2019] [Indexed: 02/07/2023] Open
Abstract
CD4+ T helper 1 (Th1) cells producing interferon gamma (IFN-γ) are critical for the resolution of visceral leishmaniasis (VL). MicroRNA 155 (miR155) promotes CD4+ Th1 responses and IFN-γ production by targeting suppressor of cytokine signaling-1 (SOCS1) and Src homology-2 domain-containing inositol 5-phosphatase 1 (SHIP-1) and therefore could play a role in the resolution of VL. To determine the role of miR155 in VL, we monitored the course of Leishmania donovani infection in miR155 knockout (miR155KO) and wild-type (WT) C57BL/6 mice. miR155KO mice displayed significantly higher liver and spleen parasite loads than WT controls and showed impaired hepatic granuloma formation. However, parasite growth eventually declined in miR155KO mice, suggesting the induction of a compensatory miR155-independent antileishmanial pathway. Leishmania antigen-stimulated splenocytes from miR155KO mice produced significantly lower levels of Th1-associated IFN-γ than controls. Interestingly, at later time points, levels of Th2-associated interleukin-4 (IL-4) and IL-10 were also lower in miR155KO splenocyte supernatants than in WT mice. On the other hand, miR155KO mice displayed significantly higher levels of IFN-γ, iNOS, and TNF-α gene transcripts in their livers than WT mice, indicating that distinct organ-specific antiparasitic mechanisms were involved in control of L. donovani infection in miR155KO mice. Throughout the course of infection, organs of miR155KO mice showed significantly more PDL1-expressing Ly6Chi inflammatory monocytes than WT mice. Conversely, blockade of Ly6Chi inflammatory monocyte recruitment in miR155KO mice significantly reduced parasitic loads, indicating that these cells contributed to disease susceptibility. In conclusion, we found that miR155 contributes to the control of L. donovani but is not essential for infection resolution.
Collapse
|
25
|
Cytokine profile and nitric oxide levels in macrophages exposed to Leishmania infantum FML. Exp Parasitol 2019; 203:1-7. [PMID: 31128104 DOI: 10.1016/j.exppara.2019.05.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 04/29/2019] [Accepted: 05/18/2019] [Indexed: 01/24/2023]
Abstract
Fucose-mannose ligand (FML) is a soluble antigen purified from Leishmania donovani complex and used for diagnosis, prognosis, and vaccine development against visceral leishmaniasis (VL). We aimed to explore the effects of FML on the production of cytokines, chemokines and nitric oxide (NO) by macrophages in vitro. Peritoneal macrophages from BALB/c mice were treated with various concentrations of FML purified from Leishmania infantum in the absence or presence of LPS Peritoneal macrophages. After 48hr, cell culture supernatants were recovered and the levels of TNF-α, IL-10, IL-12p70 and IP-10 measured by Sandwich ELISA and NO concentration by Griess reaction. We found that FML significantly increase NO, IL-12p70 and IP-10 production in both LPS-treated and untreated macrophages and increase IL-10 levels only in LPS-treated macrophages. However, FML could not alert TNF-α levels in both LPS-treated and untreated macrophages. Further analysis revealed that FML can also increase IL-12p70/IL-10 ratio in LPS-treated macrophages. We concluded that FML can polarize macrophages to an appropriate phenotype similar to M1 phenotype against Leishmania donovani complex, although IL10 and TNF results are controversial.
Collapse
|
26
|
Niu F, Liao K, Hu G, Sil S, Callen S, Guo ML, Yang L, Buch S. Cocaine-induced release of CXCL10 from pericytes regulates monocyte transmigration into the CNS. J Cell Biol 2019; 218:700-721. [PMID: 30626719 PMCID: PMC6363463 DOI: 10.1083/jcb.201712011] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 08/28/2018] [Accepted: 11/08/2018] [Indexed: 12/15/2022] Open
Abstract
Cocaine is known to facilitate the transmigration of inflammatory leukocytes into the brain, an important mechanism underlying neuroinflammation. Pericytes are well-recognized as important constituents of the blood-brain barrier (BBB), playing a key role in maintaining barrier integrity. In the present study, we demonstrate for the first time that exposure of human brain vascular pericytes to cocaine results in enhanced secretion of CXCL10, leading, in turn, to increased monocyte transmigration across the BBB both in vitro and in vivo. This process involved translocation of σ-1 receptor (σ-1R) and interaction of σ-1R with c-Src kinase, leading to activation of the Src-PDGFR-β-NF-κB pathway. These findings imply a novel role for pericytes as a source of CXCL10 in the pericyte-monocyte cross talk in cocaine-mediated neuroinflammation, underpinning their role as active components of the innate immune responses.
Collapse
Affiliation(s)
- Fang Niu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE
| | - Ke Liao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE
| | - Susmita Sil
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE
| | - Shannon Callen
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE
| | - Ming-Lei Guo
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE
| | - Lu Yang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
27
|
Masoudzadeh N, Mizbani A, Taslimi Y, Mashayekhi V, Mortazavi H, Sadeghipour P, Ardekani HM, Rafati S. Leishmania tropica infected human lesions: Whole genome transcription profiling. Acta Trop 2017; 176:236-241. [PMID: 28842129 DOI: 10.1016/j.actatropica.2017.08.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 06/24/2017] [Accepted: 08/17/2017] [Indexed: 12/22/2022]
Abstract
Leishmania (L.) tropica is the main causative agent of anthroponotic cutaneous leishmaniasis (CL) in Iran. Defining the host inflammatory response in the L. tropica lesions are crucial for the development of new treatment modalities. High-throughput RNA sequencing provides a powerful method for characterization of the human gene expression profile in L. tropica lesions. Comparing the transcription profile of the L. tropica skin lesions with normal skin identified over 5000 differentially regulated genes. Gene set enrichment analysis indicated significant activation of key immunological pathways related to antigen processing and presentation. In addition, we observed a substantial upregulation of immunoglobulin genes in lesion samples, highlighting the remarkable involvement of B cells in the infection site. To our knowledge, this study is the first report to build a comprehensive picture of transcriptome changes in acute human skin lesions during infection by L. tropica.
Collapse
Affiliation(s)
- Nasrin Masoudzadeh
- Immunotherpy and Leishmania Vaccine Research Dept., Pasteur Institute of Iran, Tehran, Iran
| | | | - Yasaman Taslimi
- Immunotherpy and Leishmania Vaccine Research Dept., Pasteur Institute of Iran, Tehran, Iran
| | - Vahid Mashayekhi
- Cutaneous Leishmaniasis Research Center, Emam Reza Hospital, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Mortazavi
- Department of Dermatology, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Pardis Sadeghipour
- Immunotherpy and Leishmania Vaccine Research Dept., Pasteur Institute of Iran, Tehran, Iran
| | | | - Sima Rafati
- Immunotherpy and Leishmania Vaccine Research Dept., Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
28
|
|
29
|
Pirdel L, Zavaran Hosseini A. Immune response to recombinant Leishmania infantum lipophosphoglycan 3 plus CpG oligodeoxynucleotides in BALB/c mice. Parasite Immunol 2017; 39. [PMID: 27353355 DOI: 10.1111/pim.12345] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 06/20/2016] [Indexed: 01/06/2023]
Abstract
Development of a protective antileishmanial vaccine is an urgent priority for successful control of different forms of leishmaniasis. The potential of a recombinant lipophosphoglycan 3 (rLPG3) expressed by Leishmania tarentolae was evaluated in combination with CpG oligodeoxynucleotides (CpG-ODN) as a Th1-promoting adjuvant against Leishmania infantum infection in BALB/c mice. First, mice were immunized subcutaneously with rLPG3 either alone or in combination with CpG-ODN. Next, the immunogenic and protective efficacies of this vaccine were analysed in immunized mice. It was observed that coadministration of rLPG3 with CpG-ODN led to enhance in a Th1 response to rLPG3 induced by itself as the IFN-γ production was promoted in association with the predominant presence of IgG2a antibodies in the sera. However, immunization with rLPG3 plus CpG-ODN induced partial protection against infectious challenge in BALB/c mice. Taken together, further studies are required to improve the protective efficacy using either more potent immune enhancers or vaccination strategies.
Collapse
Affiliation(s)
- L Pirdel
- Department of Medical Sciences, Ardabil Branch, Islamic Azad University, Ardabil, Iran
| | - A Zavaran Hosseini
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
30
|
Souza DM, Alves PM, Silva MLF, Paulino TP, Coraspe HO, Mendonça MMS, Ribeiro BM, da Silva MV, Rodrigues Júnior V, Rodrigues DBR. 5-ALA-mediated photodynamic therapy reduces the parasite load in mice infected withLeishmania braziliensis. Parasite Immunol 2017; 39. [DOI: 10.1111/pim.12403] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 11/17/2016] [Indexed: 11/30/2022]
Affiliation(s)
- D. M. Souza
- Immunology Laboratory; Department of Biological Sciences; Federal University of Triângulo Mineiro; Uberaba Minas Gerais Brazil
| | - P. M. Alves
- Immunology Laboratory; Department of Biological Sciences; Federal University of Triângulo Mineiro; Uberaba Minas Gerais Brazil
| | - M. L. F. Silva
- Immunology Laboratory; Department of Biological Sciences; Federal University of Triângulo Mineiro; Uberaba Minas Gerais Brazil
| | - T. P. Paulino
- Cefores - Centro de Educação Profissional; Federal University of Triângulo Mineiro; Uberaba Minas Gerais Brazil
| | - H. O. Coraspe
- Immunology Laboratory; Department of Biological Sciences; Federal University of Triângulo Mineiro; Uberaba Minas Gerais Brazil
| | - M. M. S. Mendonça
- Immunology Laboratory; Department of Biological Sciences; Federal University of Triângulo Mineiro; Uberaba Minas Gerais Brazil
| | - B. M. Ribeiro
- Immunology Laboratory; Department of Biological Sciences; Federal University of Triângulo Mineiro; Uberaba Minas Gerais Brazil
| | - M. V. da Silva
- Immunology Laboratory; Department of Biological Sciences; Federal University of Triângulo Mineiro; Uberaba Minas Gerais Brazil
| | - V. Rodrigues Júnior
- Immunology Laboratory; Department of Biological Sciences; Federal University of Triângulo Mineiro; Uberaba Minas Gerais Brazil
| | - D. B. R. Rodrigues
- Immunology Laboratory; Department of Biological Sciences; Federal University of Triângulo Mineiro; Uberaba Minas Gerais Brazil
- Cefores - Centro de Educação Profissional; Federal University of Triângulo Mineiro; Uberaba Minas Gerais Brazil
- Department of Immunology and Molecular Biology; University of Uberaba; Uberaba Minas Gerais Brazil
| |
Collapse
|
31
|
Davis RE, Sharma S, Conceição J, Carneiro P, Novais F, Scott P, Sundar S, Bacellar O, Carvalho EM, Wilson ME. Phenotypic and functional characteristics of HLA-DR + neutrophils in Brazilians with cutaneous leishmaniasis. J Leukoc Biol 2016; 101:739-749. [PMID: 28076241 DOI: 10.1189/jlb.4a0915-442rr] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Revised: 09/08/2016] [Accepted: 09/25/2016] [Indexed: 11/24/2022] Open
Abstract
The protozoan Leishmania braziliensis causes cutaneous leishmaniasis (CL) in endemic regions. In murine models, neutrophils (PMNs) are recruited to the site of infection soon after parasite inoculation. However, the roles of neutrophils during chronic infection and in human disease remain undefined. We hypothesized that neutrophils help maintain a systemic inflammatory state in subjects with CL. Lesion biopsies from all patients with CL tested contained neutrophils expressing HLA-DR, a molecule thought to be restricted to professional antigen-presenting cells. Although CL is a localized disease, a subset of patients with CL also had circulating neutrophils expressing HLA-DR and the costimulatory molecules CD80, CD86, and CD40. PMNs isolated from a low-density leukocyte blood fraction (LD-PMNs) contained a higher percentage of HLA-DR+ PMNs than did normal-density PMNs. In vitro coculture experiments suggested LD-PMNs do not suppress T cell responses, differentiating them from MDSCs. Flow-sorted HLA-DR+ PMNs morphologically resembled conventional PMNs, and they exhibited functional properties of PMNs. Compared with conventional PMNs, HLA-DR+ PMNs showed increased activation, degranulation, DHR123 oxidation, and phagocytic capacity. A few HLA-DR+ PMNs were observed in healthy subjects, and that proportion could be increased by incubation in either inflammatory cytokines or in plasma from a patient with CL. This was accompanied by an increase in PMN hladrb1 mRNA, suggesting a possible connection between neutrophil "priming" and up-regulation of HLA-DR. These data suggest that PMNs that are primed for activation and that also express surface markers of antigen-presenting cells emerge in the circulation and infected tissue lesions of patients with CL.
Collapse
Affiliation(s)
- Richard E Davis
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Smriti Sharma
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Jacilara Conceição
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil.,Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais, INCT-DT (CNPq/MCT), Salvador, Bahia, Brazil
| | - Pedro Carneiro
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil.,Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais, INCT-DT (CNPq/MCT), Salvador, Bahia, Brazil
| | - Fernanda Novais
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shyam Sundar
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Olivia Bacellar
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil.,Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais, INCT-DT (CNPq/MCT), Salvador, Bahia, Brazil
| | - Edgar M Carvalho
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil.,Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais, INCT-DT (CNPq/MCT), Salvador, Bahia, Brazil.,Fundação Gonçalo Muniz, Fiocruz-Bahia, Salvador, Bahia Brazil
| | - Mary E Wilson
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA; .,Departments of Internal Medicine and Microbiology, University of Iowa, Iowa City, Iowa, USA; and.,Research Service, Iowa City Veterans' Affairs Medical Center, Iowa City, Iowa, USA
| |
Collapse
|
32
|
Rutar M, Natoli R, Chia RX, Valter K, Provis JM. Chemokine-mediated inflammation in the degenerating retina is coordinated by Müller cells, activated microglia, and retinal pigment epithelium. J Neuroinflammation 2015; 12:8. [PMID: 25595590 PMCID: PMC4308937 DOI: 10.1186/s12974-014-0224-1] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/18/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Monocyte infiltration is involved in the pathogenesis of many retinal degenerative conditions. This process traditionally depends on local expression of chemokines, though the roles of many of these in the degenerating retina are unclear. Here, we investigate expression and in situ localization of the broad chemokine response in a light-induced model of retinal degeneration. METHODS Sprague-Dawley (SD) rats were exposed to 1,000 lux light damage (LD) for up to 24 hrs. At time points during (1 to 24 hrs) and following (3 and 7 days) exposure, animals were euthanized and retinas processed. Microarray analysis assessed differential expression of chemokines. Some genes were further investigated using polymerase chain reaction (PCR) and in situ hybridization and contrasted with photoreceptor apoptosis using terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL). Recruitment of retinal CD45 (+) leukocytes was determined via fluorescence activated cell sorting (FACS), and expression of chemokine receptors determined using PCR. RESULTS Exposure to 24 hrs of LD resulted in differential expression of chemokines including Ccl3, Ccl4, Ccl7, Cxcl1, and Cxcl10. Their upregulation correlated strongly with peak photoreceptor death, at 24 hrs exposure. In situ hybridization revealed that the modulated chemokines were expressed by a combination of Müller cells, activated microglia, and retinal pigment epithelium (RPE). This preceded large increases in the number of CD45(+) cells at 3- and 7-days post exposure, which expressed a corresponding repertoire of chemokine receptors. CONCLUSIONS Our data indicate that retinal degeneration induces upregulation of a broad chemokine response whose expression is coordinated by Müller cells, microglia, and RPE. The findings inform our understanding of the processes govern the trafficking of leukocytes, which are contributors in the pathology of retinal degenerations.
Collapse
Affiliation(s)
- Matt Rutar
- John Curtin School of Medical Research, The Australian National University, Building 131, Garran Road, Canberra, ACT 2601, Australia. .,ANU Medical School, The Australian National University, 54 Mills Road, Canberra, ACT 2601, Australia.
| | - Riccardo Natoli
- John Curtin School of Medical Research, The Australian National University, Building 131, Garran Road, Canberra, ACT 2601, Australia. .,ANU Medical School, The Australian National University, 54 Mills Road, Canberra, ACT 2601, Australia.
| | - R X Chia
- John Curtin School of Medical Research, The Australian National University, Building 131, Garran Road, Canberra, ACT 2601, Australia.
| | - Krisztina Valter
- John Curtin School of Medical Research, The Australian National University, Building 131, Garran Road, Canberra, ACT 2601, Australia. .,ANU Medical School, The Australian National University, 54 Mills Road, Canberra, ACT 2601, Australia.
| | - Jan M Provis
- John Curtin School of Medical Research, The Australian National University, Building 131, Garran Road, Canberra, ACT 2601, Australia. .,ANU Medical School, The Australian National University, 54 Mills Road, Canberra, ACT 2601, Australia.
| |
Collapse
|
33
|
Smirnova NF, Gayral S, Pedros C, Loirand G, Vaillant N, Malet N, Kassem S, Calise D, Goudounèche D, Wymann MP, Hirsch E, Gadeau AP, Martinez LO, Saoudi A, Laffargue M. Targeting PI3Kγ activity decreases vascular trauma-induced intimal hyperplasia through modulation of the Th1 response. ACTA ACUST UNITED AC 2014; 211:1779-92. [PMID: 25073791 PMCID: PMC4144742 DOI: 10.1084/jem.20131276] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Interventional strategies to treat atherosclerosis, such as transluminal angioplasty and stent implantation, often cause vascular injury. This leads to intimal hyperplasia (IH) formation that induces inflammatory and fibroproliferative processes and ultimately restenosis. We show that phosphoinositide 3-kinase γ (PI3Kγ) is a key player in IH formation and is a valid therapeutic target in its prevention/treatment. PI3Kγ-deficient mice and mice expressing catalytically inactive PI3Kγ (PI3Kγ KD) showed reduced arterial occlusion and accumulation of monocytes and T cells around sites of vascular lesion. The transfer of PI3Kγ KD CD4(+) T cells into Rag2-deficient mice greatly reduced vascular occlusion compared with WT cells, clearly demonstrating the involvement of PI3Kγ in CD4(+) T cells during IH formation. In addition we found that IH is associated with increased levels of Th1 and Th17 cytokines. A specific decrease in the Th1 response was observed in the absence of PI3Kγ activity, leading to decreased CXCL10 and RANTES production by smooth muscle cells. Finally, we show that treatment with the PI3Kγ inhibitor AS-605240 is sufficient to decrease IH in both mouse and rat models, reinforcing the therapeutic potential of PI3Kγ inhibition. Altogether, these findings demonstrate a new role for PI3Kγ activity in Th1-controlled IH development.
Collapse
Affiliation(s)
- Natalia F Smirnova
- INSERM, UMR1048, F-31300 Toulouse, France Université Toulouse III, Institut de Maladies Métaboliques et Cardiovasculaires, F-31300 Toulouse, France
| | - Stéphanie Gayral
- INSERM, UMR1048, F-31300 Toulouse, France Université Toulouse III, Institut de Maladies Métaboliques et Cardiovasculaires, F-31300 Toulouse, France
| | - Christophe Pedros
- INSERM, UMR1043, F-31300 Toulouse, France UMR CNRS, U5282, F-31300 Toulouse, France Université de Toulouse, UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP), F-31300 Toulouse, France
| | - Gervaise Loirand
- INSERM, UMR1087, F-44007 Nantes, France CNRS 6291, F-44007 Nantes, France
| | - Nathalie Vaillant
- INSERM, UMR1087, F-44007 Nantes, France CNRS 6291, F-44007 Nantes, France
| | - Nicole Malet
- INSERM, UMR1048, F-31300 Toulouse, France Université Toulouse III, Institut de Maladies Métaboliques et Cardiovasculaires, F-31300 Toulouse, France
| | - Sahar Kassem
- INSERM, UMR1043, F-31300 Toulouse, France UMR CNRS, U5282, F-31300 Toulouse, France Université de Toulouse, UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP), F-31300 Toulouse, France
| | - Denis Calise
- INSERM, UMR1048, F-31300 Toulouse, France Université Toulouse III, Institut de Maladies Métaboliques et Cardiovasculaires, F-31300 Toulouse, France
| | - Dominique Goudounèche
- Université Toulouse III, Institut de Maladies Métaboliques et Cardiovasculaires, F-31300 Toulouse, France CMEAB, F-31000 Toulouse, France
| | - Matthias P Wymann
- Institute of Biochemistry and Genetics, University of Basel, 4058 Basel, Switzerland
| | - Emilio Hirsch
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Turin, 10124 Turin, Italy
| | | | - Laurent O Martinez
- INSERM, UMR1048, F-31300 Toulouse, France Université Toulouse III, Institut de Maladies Métaboliques et Cardiovasculaires, F-31300 Toulouse, France
| | - Abdelhadi Saoudi
- INSERM, UMR1043, F-31300 Toulouse, France UMR CNRS, U5282, F-31300 Toulouse, France Université de Toulouse, UPS, Centre de Physiopathologie de Toulouse Purpan (CPTP), F-31300 Toulouse, France
| | - Muriel Laffargue
- INSERM, UMR1048, F-31300 Toulouse, France Université Toulouse III, Institut de Maladies Métaboliques et Cardiovasculaires, F-31300 Toulouse, France
| |
Collapse
|
34
|
Novais FO, Carvalho LP, Passos S, Roos DS, Carvalho EM, Scott P, Beiting DP. Genomic profiling of human Leishmania braziliensis lesions identifies transcriptional modules associated with cutaneous immunopathology. J Invest Dermatol 2014; 135:94-101. [PMID: 25036052 PMCID: PMC4268311 DOI: 10.1038/jid.2014.305] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 04/23/2014] [Accepted: 05/06/2014] [Indexed: 11/24/2022]
Abstract
The host immune response plays a critical role not only in protection from human leishmaniasis, but also in promoting disease severity. Although candidate gene approaches in mouse models of leishmaniasis have been extremely informative, a global understanding of the immune pathways active in lesions from human patients is lacking. To address this issue, genome-wide transcriptional profiling of Leishmania braziliensis-infected cutaneous lesions and normal skin controls was carried out. A signature of the L. braziliensis skin lesion was defined that includes over 2,000 differentially regulated genes. Pathway-level analysis of this transcriptional response revealed key biological pathways present in cutaneous lesions, generating a testable ‘metapathway’ model of immunopathology, and providing new insights for treatment of human leishmaniasis.
Collapse
Affiliation(s)
- Fernanda O Novais
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lucas P Carvalho
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais-INCT-DT(CNPq/MCT), Serviço de Imunologia, Hospital Universitario Prof. Edgard Santos, Universidade Federal da Bahia Salvador, Bahia, Brazil
| | - Sara Passos
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais-INCT-DT(CNPq/MCT), Serviço de Imunologia, Hospital Universitario Prof. Edgard Santos, Universidade Federal da Bahia Salvador, Bahia, Brazil
| | - David S Roos
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edgar M Carvalho
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais-INCT-DT(CNPq/MCT), Serviço de Imunologia, Hospital Universitario Prof. Edgard Santos, Universidade Federal da Bahia Salvador, Bahia, Brazil
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | - Daniel P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
35
|
Hartley MA, Drexler S, Ronet C, Beverley SM, Fasel N. The immunological, environmental, and phylogenetic perpetrators of metastatic leishmaniasis. Trends Parasitol 2014; 30:412-22. [PMID: 24954794 DOI: 10.1016/j.pt.2014.05.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 05/23/2014] [Accepted: 05/24/2014] [Indexed: 02/07/2023]
Abstract
Cutaneous leishmaniases have persisted for centuries as chronically disfiguring parasitic infections affecting millions of people across the subtropics. Symptoms range from the more prevalent single, self-healing cutaneous lesion to a persistent, metastatic disease, where ulcerations and granulomatous nodules can affect multiple secondary sites of the skin and delicate facial mucosa, even sometimes diffusing throughout the cutaneous system as a papular rash. The basis for such diverse pathologies is multifactorial, ranging from parasite phylogeny to host immunocompetence and various environmental factors. Although complex, these pathologies often prey on weaknesses in the innate immune system and its pattern recognition receptors. This review explores the observed and potential associations among the multifactorial perpetrators of infectious metastasis and components of the innate immune system.
Collapse
Affiliation(s)
- Mary-Anne Hartley
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Stefan Drexler
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Catherine Ronet
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Stephen M Beverley
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nicolas Fasel
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland.
| |
Collapse
|
36
|
Ives A, Masina S, Castiglioni P, Prével F, Revaz-Breton M, Hartley MA, Launois P, Fasel N, Ronet C. MyD88 and TLR9 dependent immune responses mediate resistance to Leishmania guyanensis infections, irrespective of Leishmania RNA virus burden. PLoS One 2014; 9:e96766. [PMID: 24801628 PMCID: PMC4011865 DOI: 10.1371/journal.pone.0096766] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 04/11/2014] [Indexed: 12/20/2022] Open
Abstract
Infections with Leishmania parasites of the Leishmania Viannia subgenus give rise to both localized cutaneous (CL), and metastatic leishmaniasis. Metastasizing disease forms including disseminated (DCL) and mutocutaneous (MCL) leishmaniasis result from parasitic dissemination and lesion formation at sites distal to infection and have increased inflammatory responses. The presence of Leishmania RNA virus (LRV) in L. guyanensis parasites contributes to the exacerbation of disease and impacts inflammatory responses via activation of TLR3 by the viral dsRNA. In this study we investigated other innate immune response adaptor protein modulators and demonstrated that both MyD88 and TLR9 played a crucial role in the development of Th1-dependent healing responses against L. guyanensis parasites regardless of their LRV status. The absence of MyD88- or TLR9-dependent signaling pathways resulted in increased Th2 associated cytokines (IL-4 and IL-13), which was correlated with low transcript levels of IL-12p40. The reliance of IL-12 was further confirmed in IL12AB−/− mice, which were completely susceptible to infection. Protection to L. guyanensis infection driven by MyD88- and TLR9-dependent immune responses arises independently to those induced due to high LRV burden within the parasites.
Collapse
Affiliation(s)
- Annette Ives
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Slavica Masina
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Patrik Castiglioni
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Florence Prével
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Mélanie Revaz-Breton
- Department of Biochemistry, World Health Organization Immunology Research and Training center (WHO-IRTC), Epalinges, Switzerland
| | - Mary-Anne Hartley
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Pascal Launois
- Department of Biochemistry, World Health Organization Immunology Research and Training center (WHO-IRTC), Epalinges, Switzerland
| | - Nicolas Fasel
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Catherine Ronet
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
- Department of Biochemistry, World Health Organization Immunology Research and Training center (WHO-IRTC), Epalinges, Switzerland
- * E-mail:
| |
Collapse
|
37
|
Duell BL, Carey AJ, Dando SJ, Schembri MA, Ulett GC. Human bladder uroepithelial cells synergize with monocytes to promote IL-10 synthesis and other cytokine responses to uropathogenic Escherichia coli. PLoS One 2013; 8:e78013. [PMID: 24155979 PMCID: PMC3796480 DOI: 10.1371/journal.pone.0078013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 09/15/2013] [Indexed: 12/31/2022] Open
Abstract
Urinary tract infections are a major source of morbidity for women and the elderly, with Uropathogenic Escherichia coli (UPEC) being the most prevalent causative pathogen. Studies in recent years have defined a key anti-inflammatory role for Interleukin-10 (IL-10) in urinary tract infection mediated by UPEC and other uropathogens. We investigated the nature of the IL-10-producing interactions between UPEC and host cells by utilising a novel co-culture model that incorporated lymphocytes, mononuclear and uroepithelial cells in histotypic proportions. This co-culture model demonstrated synergistic IL-10 production effects between monocytes and uroepithelial cells following infection with UPEC. Membrane inserts were used to separate the monocyte and uroepithelial cell types during infection and revealed two synergistic IL-10 production effects based on contact-dependent and soluble interactions. Analysis of a comprehensive set of immunologically relevant biomarkers in monocyte-uroepithelial cell co-cultures highlighted that multiple cytokine, chemokine and signalling factors were also produced in a synergistic or antagonistic fashion. These results demonstrate that IL-10 responses to UPEC occur via multiple interactions between several cells types, implying a complex role for infection-related IL-10 during UTI. Development and application of the co-culture model described in this study is thus useful to define the degree of contact dependency of biomarker production to UPEC, and highlights the relevance of histotypic co-cultures in studying complex host-pathogen interactions.
Collapse
Affiliation(s)
- Benjamin L. Duell
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| | - Alison J. Carey
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| | - Samantha J. Dando
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Mark A. Schembri
- School of Chemical and Molecular Biosciences, University of Queensland, Brisbane, Queensland, Australia
| | - Glen C. Ulett
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
- * E-mail:
| |
Collapse
|
38
|
Lemaire J, Mkannez G, Guerfali FZ, Gustin C, Attia H, Sghaier RM, Sysco-Consortium, Dellagi K, Laouini D, Renard P. MicroRNA expression profile in human macrophages in response to Leishmania major infection. PLoS Negl Trop Dis 2013; 7:e2478. [PMID: 24098824 PMCID: PMC3789763 DOI: 10.1371/journal.pntd.0002478] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 08/30/2013] [Indexed: 12/31/2022] Open
Abstract
Background Leishmania (L.) are intracellular protozoan parasites able to survive and replicate in the hostile phagolysosomal environment of infected macrophages. They cause leishmaniasis, a heterogeneous group of worldwide-distributed affections, representing a paradigm of neglected diseases that are mainly embedded in impoverished populations. To establish successful infection and ensure their own survival, Leishmania have developed sophisticated strategies to subvert the host macrophage responses. Despite a wealth of gained crucial information, these strategies still remain poorly understood. MicroRNAs (miRNAs), an evolutionarily conserved class of endogenous 22-nucleotide non-coding RNAs, are described to participate in the regulation of almost every cellular process investigated so far. They regulate the expression of target genes both at the levels of mRNA stability and translation; changes in their expression have a profound effect on their target transcripts. Methodology/Principal Findings We report in this study a comprehensive analysis of miRNA expression profiles in L. major-infected human primary macrophages of three healthy donors assessed at different time-points post-infection (three to 24 h). We show that expression of 64 out of 365 analyzed miRNAs was consistently deregulated upon infection with the same trends in all donors. Among these, several are known to be induced by TLR-dependent responses. GO enrichment analysis of experimentally validated miRNA-targeted genes revealed that several pathways and molecular functions were disturbed upon parasite infection. Finally, following parasite infection, miR-210 abundance was enhanced in HIF-1α-dependent manner, though it did not contribute to inhibiting anti-apoptotic pathways through pro-apoptotic caspase-3 regulation. Conclusions/Significance Our data suggest that alteration in miRNA levels likely plays an important role in regulating macrophage functions following L. major infection. These results could contribute to better understanding of the dynamics of gene expression in host cells during leishmaniasis. Leishmania parasites belong to different species, each one characterized by specific vectors and reservoirs, and causes cutaneous or visceral disease(s) of variable clinical presentation and severity. In its mammalian host, the parasite is an obligate intracellular pathogen infecting the monocyte/macrophage lineage. Leishmania have developed ambiguous relationships with macrophages. Indeed, these cells are the shelter of invading parasites, where they will grow and eventually will reside in a silent state for life. But macrophages are also the cells that participate, through the induction of several pro-inflammatory mediators and antigen presentation, to shape the host immune response and ultimately kill the invader. To subvert these anti-parasite responses, Leishmania manipulate the host machinery for their own differentiation and survival. We aimed to evaluate the impact of L. major (the causative agent of zoonotic cutaneous leishmaniasis) infection on deregulation of non-coding miRNAs, a class of important regulators of gene expression. Our results revealed the implication of several miRNAs on macrophage fate upon parasite infection through regulation of different pathways, including cell death. Our findings provided a new insight for understanding mechanisms governing this miRNA deregulation by parasite infection and will help to provide clues for the development of control strategies for this disease.
Collapse
Affiliation(s)
- Julien Lemaire
- Laboratory of Biochemistry and Cellular Biology (URBC), NARILIS-University of Namur, Namur, Belgium
| | - Ghada Mkannez
- Institut Pasteur de Tunis, LR11IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
| | - Fatma Z. Guerfali
- Institut Pasteur de Tunis, LR11IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
| | - Cindy Gustin
- Laboratory of Biochemistry and Cellular Biology (URBC), NARILIS-University of Namur, Namur, Belgium
| | - Hanène Attia
- Institut Pasteur de Tunis, LR11IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
| | - Rabiaa M. Sghaier
- Institut Pasteur de Tunis, LR11IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
| | | | - Koussay Dellagi
- Institut Pasteur de Tunis, LR11IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
- Institut de Recherche pour le Développement (IRD) et Centre de Recherche et de Veille sur les Maladies Emergentes dans l'Océan Indien (CRVOI), Sainte Clotilde, Reunion Island, France
| | - Dhafer Laouini
- Institut Pasteur de Tunis, LR11IPT02, Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), Tunis-Belvédère, Tunisia
- Université Tunis El Manar, Tunis, Tunisia
- * E-mail: , (DL); (PR)
| | - Patricia Renard
- Laboratory of Biochemistry and Cellular Biology (URBC), NARILIS-University of Namur, Namur, Belgium
- * E-mail: , (DL); (PR)
| |
Collapse
|
39
|
Abstract
Leishmaniasis is a complex disease that is caused by parasites of the Leishmania genus. Leishmania are further classified into several complexes, each of which can engage in distinct interactions with mammalian hosts resulting in differing disease presentations. It is therefore not unexpected that host immune responses to Leishmania are variable. The induction of interferon gamma (IFN-γ) and response to it in these infections has received considerable attention. In this review, we summarize our current understanding of some of the host responses during Leishmania infections that are regulated by IFN-γ. In addition, studies that explore the nature of parasite-derived molecular mediators that might affect the host response to IFN-γ are also discussed.
Collapse
Affiliation(s)
- Peter E Kima
- Department of Microbiology and Cell Science, University of Florida , Gainesville, FL , USA
| | | |
Collapse
|
40
|
Diaz NL, Zerpa O, Tapia FJ. Chemokines and chemokine receptors expression in the lesions of patients with American cutaneous leishmaniasis. Mem Inst Oswaldo Cruz 2013; 108:446-52. [DOI: 10.1590/s0074-0276108042013008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 03/27/2013] [Indexed: 12/20/2022] Open
|
41
|
Díaz NL, Zerpa O, Tapia FJ. Chemokines and chemokine receptors expression in the lesions of patients with American cutaneous leishmaniasis. Mem Inst Oswaldo Cruz 2013; 108. [PMID: 23827991 PMCID: PMC3970620 DOI: 10.1590/0074-0276108042013008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
American cutaneous leishmaniasis (ACL) presents distinct active clinical forms with different grades of severity, known as localised (LCL), intermediate (ICL) and diffuse (DCL) cutaneous leishmaniasis. LCL and DCL are associated with a polarised T-helper (Th)1 and Th2 immune response, respectively, whereas ICL, or chronic cutaneous leishmaniasis, is associated with an exacerbated immune response and a mixed cytokine expression profile. Chemokines and chemokine receptors are involved in cellular migration and are critical in the inflammatory response. Therefore, we evaluated the expression of the chemokines CXCL10, CCL4, CCL8, CCL11 and CXCL8 and the chemokine receptors CCR3, CXCR3, CCR5 and CCR7 in the lesions of patients with different clinical forms of ACL using immunohistochemistry. LCL patients exhibited a high density of CXCL10+, CCL4+ and CCL8+ cells, indicating an important role for these chemokines in the local Th1 immune response and the migration of CXCR3+ cells. LCL patients showed a higher density of CCR7+ cells than ICL or DCL patients, suggesting major dendritic cell (DC) migration to lymph nodes. Furthermore, DCL was associated with low expression levels of Th1-associated chemokines and CCL11+ epidermal DCs, which contribute to the recruitment of CCR3+ cells. Our findings also suggest an important role for epidermal cells in the induction of skin immune responses through the production of chemokines, such as CXCL10, by keratinocytes.
Collapse
Affiliation(s)
- Nilka Luisa Díaz
- Instituto de Biomedicina, Universidad Central de Venezuela, Caracas,
Venezuela , Corresponding author:
| | - Olga Zerpa
- Instituto de Biomedicina, Universidad Central de Venezuela, Caracas,
Venezuela
| | - Félix Jacobo Tapia
- Instituto de Biomedicina, Universidad Central de Venezuela, Caracas,
Venezuela
| |
Collapse
|
42
|
Björkander S, Heidari-Hamedani G, Bremme K, Gunnarsson I, Holmlund U. Peripheral Monocyte Expression of the Chemokine Receptors CCR2, CCR5 and CXCR3 is Altered at Parturition in Healthy Women and in Women with Systemic Lupus Erythematosus. Scand J Immunol 2013; 77:200-12. [DOI: 10.1111/sji.12021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 12/18/2012] [Indexed: 12/22/2022]
Affiliation(s)
- S. Björkander
- Department of Molecular Biosciences; The Wenner-Gren Institute; Stockholm University; Stockholm; Sweden
| | - G. Heidari-Hamedani
- Department of Molecular Biosciences; The Wenner-Gren Institute; Stockholm University; Stockholm; Sweden
| | - K. Bremme
- Division of Obstetrics and Gynecology; Department of Women and Child Health; Karolinska University Hospital; Karolinska Institute; Stockholm; Sweden
| | - I. Gunnarsson
- Department of Medicine; Unit of Rheumatology; Karolinska University Hospital; Karolinska Institute; Stockholm; Sweden
| | - U. Holmlund
- Department of Molecular Biosciences; The Wenner-Gren Institute; Stockholm University; Stockholm; Sweden
| |
Collapse
|
43
|
Zangger H, Ronet C, Desponds C, Kuhlmann FM, Robinson J, Hartley MA, Prevel F, Castiglioni P, Pratlong F, Bastien P, Müller N, Parmentier L, Saravia NG, Beverley SM, Fasel N. Detection of Leishmania RNA virus in Leishmania parasites. PLoS Negl Trop Dis 2013; 7:e2006. [PMID: 23326619 PMCID: PMC3542153 DOI: 10.1371/journal.pntd.0002006] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 11/28/2012] [Indexed: 12/22/2022] Open
Abstract
Background Patients suffering from cutaneous leishmaniasis (CL) caused by New World Leishmania (Viannia) species are at high risk of developing mucosal (ML) or disseminated cutaneous leishmaniasis (DCL). After the formation of a primary skin lesion at the site of the bite by a Leishmania-infected sand fly, the infection can disseminate to form secondary lesions. This metastatic phenotype causes significant morbidity and is often associated with a hyper-inflammatory immune response leading to the destruction of nasopharyngeal tissues in ML, and appearance of nodules or numerous ulcerated skin lesions in DCL. Recently, we connected this aggressive phenotype to the presence of Leishmania RNA virus (LRV) in strains of L. guyanensis, showing that LRV is responsible for elevated parasitaemia, destructive hyper-inflammation and an overall exacerbation of the disease. Further studies of this relationship and the distribution of LRVs in other Leishmania strains and species would benefit from improved methods of viral detection and quantitation, especially ones not dependent on prior knowledge of the viral sequence as LRVs show significant evolutionary divergence. Methodology/Principal Findings This study reports various techniques, among which, the use of an anti-dsRNA monoclonal antibody (J2) stands out for its specific and quantitative recognition of dsRNA in a sequence-independent fashion. Applications of J2 include immunofluorescence, ELISA and dot blot: techniques complementing an arsenal of other detection tools, such as nucleic acid purification and quantitative real-time-PCR. We evaluate each method as well as demonstrate a successful LRV detection by the J2 antibody in several parasite strains, a freshly isolated patient sample and lesion biopsies of infected mice. Conclusions/Significance We propose that refinements of these methods could be transferred to the field for use as a diagnostic tool in detecting the presence of LRV, and potentially assessing the LRV-related risk of complications in cutaneous leishmaniasis. The endosymbiosis of viruses in microbes is a well-described and prevalent environmental partnership, where viruses offer their cellular host incentives of fitness in exchange for the use of their metabolic machinery. We have recently exposed this as an important factor in certain metastatic leishmaniases of South America, where the nucleic acid of a virus residing within some Leishmania parasites acts as a potent innate immunogen causing a destructive inflammatory response, which worsens disease. Leishmania RNA Virus (LRV) exists within many species of Leishmania as a stable infection; these LRV positive strains have been found throughout South America in cutaneous leishmaniases that are often complicated by the occurrence of infectious metastasis with an underlying hyperinflammatory response. In this report, we describe the use of an anti-dsRNA monoclonal antibody (J2), which specifically recognizes dsRNA in a quantitative and sequence-independent fashion. Refined versions of these methods could be transferred to the field as diagnostic tools for detecting the presence of LRV (or other dsRNA viruses), and potentially assessing the LRV-related risk of complicated cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Haroun Zangger
- Department of Biochemistry, University of Lausanne, Epalinges, Vaud, Switzerland
| | - Catherine Ronet
- Department of Biochemistry, University of Lausanne, Epalinges, Vaud, Switzerland
| | - Chantal Desponds
- Department of Biochemistry, University of Lausanne, Epalinges, Vaud, Switzerland
| | - F. Matthew Kuhlmann
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - John Robinson
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Mary-Anne Hartley
- Department of Biochemistry, University of Lausanne, Epalinges, Vaud, Switzerland
| | - Florence Prevel
- Department of Biochemistry, University of Lausanne, Epalinges, Vaud, Switzerland
| | - Patrik Castiglioni
- Department of Biochemistry, University of Lausanne, Epalinges, Vaud, Switzerland
| | - Francine Pratlong
- French National Reference Centre for Leishmaniases, Département de Parasitologie-Mycologie, CHRU de Montpellier, Montpellier, France
- University Montpellier 1, Faculty of Medicine, UMR CNRS 5290/IRD 224/UM1/UM2 “MIVEGEC”, Montpellier, France
| | - Patrick Bastien
- French National Reference Centre for Leishmaniases, Département de Parasitologie-Mycologie, CHRU de Montpellier, Montpellier, France
- University Montpellier 1, Faculty of Medicine, UMR CNRS 5290/IRD 224/UM1/UM2 “MIVEGEC”, Montpellier, France
| | - Norbert Müller
- Institute of Parasitology, Vetsuisse Faculty Berne, University of Bern, Bern, Switzerland
| | - Laurent Parmentier
- Department of Dermatology, Hôpitaux du Valais, Sierre, Valais, Switzerland
| | - Nancy Gore Saravia
- Centro Internacional de Entrenamiento e Investigaciones Médicas (CIDEIM), Cali, Colombia
| | - Stephen M. Beverley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Nicolas Fasel
- Department of Biochemistry, University of Lausanne, Epalinges, Vaud, Switzerland
- * E-mail:
| |
Collapse
|
44
|
Soong L, Henard CA, Melby PC. Immunopathogenesis of non-healing American cutaneous leishmaniasis and progressive visceral leishmaniasis. Semin Immunopathol 2012; 34:735-51. [PMID: 23053396 PMCID: PMC4111229 DOI: 10.1007/s00281-012-0350-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 09/21/2012] [Indexed: 12/21/2022]
Abstract
The outcomes of Leishmania infection are determined by host immune and nutrition status, parasite species, and co-infection with other pathogens. While subclinical infection and self-healing cutaneous leishmaniasis (CL) are common, uncontrolled parasite replication can lead to non-healing local lesions or visceral leishmaniasis (VL). It is known that infection control requires Th1-differentiation cytokines (IL-12, IL-18, and IL-27) and Th1 cell and macrophage activation. However, there is no generalized consensus for the mechanisms of host susceptibility. The recent studies on regulatory T cells and IL-17-producing cells help explain the effector T cell responses that occur independently of the known Th1/Th2 cell signaling pathways. This review focuses on the immunopathogenesis of non-healing American CL and progressive VL. We summarize recent evidence from human and animal studies that reveals the mechanisms of dysregulated, hyper-responses to Leishmania braziliensis, as well as the presence of disease-promoting or the absence of protective responses to Leishmania amazonensis and Leishmania donovani. We highlight immune-mediated parasite growth and immunopathogenesis, with an emphasis on the putative roles of IL-17 and its related cytokines as well as arginase. A better understanding of the quality and regulation of innate immunity and T cell responses triggered by Leishmania will aid in the rational control of pathology and the infection.
Collapse
Affiliation(s)
- Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555-1070, USA.
| | | | | |
Collapse
|
45
|
Parise-Filho R, Pasqualoto KFM, Magri FMM, Ferreira AK, da Silva BAVG, Damião MCFCB, Tavares MT, Azevedo RA, Auada AVV, Polli MC, Brandt CA. Dillapiole as antileishmanial agent: discovery, cytotoxic activity and preliminary SAR studies of dillapiole analogues. Arch Pharm (Weinheim) 2012; 345:934-44. [PMID: 22996811 DOI: 10.1002/ardp.201200212] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 07/20/2012] [Accepted: 07/24/2012] [Indexed: 11/06/2022]
Abstract
In this paper, the isolation of dillapiole (1) from Piper aduncum was reported as well as the semi-synthesis of two phenylpropanoid derivatives [di-hydrodillapiole (2), isodillapiole (3)], via reduction and isomerization reactions. Also, the compounds' molecular properties (structural, electronic, hydrophobic, and steric) were calculated and investigated to establish some preliminary structure-activity relationships (SAR). Compounds were evaluated for in vitro antileishmanial activity and cytotoxic effects on fibroblast cells. Compound 1 presented inhibitory activity against Leishmania amazonensis (IC(50) = 69.3 µM) and Leishmania brasiliensis (IC(50) = 59.4 µM) and induced cytotoxic effects on fibroblast cells mainly in high concentrations. Compounds 2 (IC(50) = 99.9 µM for L. amazonensis and IC(50) = 90.5 µM for L. braziliensis) and 3 (IC(50) = 122.9 µM for L. amazonensis and IC(50) = 109.8 µM for L. brasiliensis) were less active than dillapiole (1). Regarding the molecular properties, the conformational arrangement of the side chain, electronic features, and the hydrophilic/hydrophobic balance seem to be relevant for explaining the antileishmanial activity of dillapiole and its analogues.
Collapse
Affiliation(s)
- Roberto Parise-Filho
- Faculty of Pharmaceutical Sciences, Department of Pharmacy, University of São Paulo, 05508-900 São Paulo, Brazil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Transcriptomic signature of Leishmania infected mice macrophages: a metabolic point of view. PLoS Negl Trop Dis 2012; 6:e1763. [PMID: 22928052 PMCID: PMC3424254 DOI: 10.1371/journal.pntd.0001763] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 06/21/2012] [Indexed: 11/23/2022] Open
Abstract
We analyzed the transcriptional signatures of mouse bone marrow-derived macrophages at different times after infection with promastigotes of the protozoan parasite Leishmania major. Ingenuity Pathway Analysis revealed that the macrophage metabolic pathways including carbohydrate and lipid metabolisms were among the most altered pathways at later time points of infection. Indeed, L. major promastiogtes induced increased mRNA levels of the glucose transporter and almost all of the genes associated with glycolysis and lactate dehydrogenase, suggesting a shift to anaerobic glycolysis. On the other hand, L. major promastigotes enhanced the expression of scavenger receptors involved in the uptake of Low-Density Lipoprotein (LDL), inhibited the expression of genes coding for proteins regulating cholesterol efflux, and induced the synthesis of triacylglycerides. These data suggested that Leishmania infection disturbs cholesterol and triglycerides homeostasis and may lead to cholesterol accumulation and foam cell formation. Using Filipin and Bodipy staining, we showed cholesterol and triglycerides accumulation in infected macrophages. Moreover, Bodipy-positive lipid droplets accumulated in close proximity to parasitophorous vacuoles, suggesting that intracellular L. major may take advantage of these organelles as high-energy substrate sources. While the effect of infection on cholesterol accumulation and lipid droplet formation was independent on parasite development, our data indicate that anaerobic glycolysis is actively induced by L. major during the establishment of infection. Leishmania are obligated intracellular pathogens that develop almost exclusively in macrophages. Experimental leishmaniasis in mice is one of the most extensively studied models of intracellular infections both at the level of the parasite and host immune responses. We took advantage of Balb/c mice model to investigate gene expression profile through Affymetrix oligonucleotide arrays. In order to have a general and dynamic picture of the complex biological events that are acting in the context of Leishmania intracellular parasitism, we investigated the mouse macrophage response to initial invasion of L. major over a time course that extended from one to 24 hours post-infection. Our results reveal the alteration of several biological processes and metabolic changes. Indeed, similarly to different other pathogens, Leishmania induces cholesterol accumulation and foam cell formation that have been confirmed by confocal microscopy experiments. Whether Leishmania parasites take advantage of this high-energy source is now under investigation. Our findings provided further understandings in host responses to Leishmania infection.
Collapse
|
47
|
Hartley MA, Ronet C, Zangger H, Beverley SM, Fasel N. Leishmania RNA virus: when the host pays the toll. Front Cell Infect Microbiol 2012; 2:99. [PMID: 22919688 PMCID: PMC3417650 DOI: 10.3389/fcimb.2012.00099] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 06/27/2012] [Indexed: 12/21/2022] Open
Abstract
The presence of an RNA virus in a South American subgenus of the Leishmania parasite, L. (Viannia), was detected several decades ago but its role in leishmanial virulence and metastasis was only recently described. In Leishmania guyanensis, the nucleic acid of Leishmania RNA virus (LRV1) acts as a potent innate immunogen, eliciting a hyper-inflammatory immune response through toll-like receptor 3 (TLR3). The resultant inflammatory cascade has been shown to increase disease severity, parasite persistence, and perhaps even resistance to anti-leishmanial drugs. Curiously, LRVs were found mostly in clinical isolates prone to infectious metastasis in both their human source and experimental animal model, suggesting an association between the viral hyperpathogen and metastatic complications such as mucocutaneous leishmaniasis (MCL). MCL presents as chronic secondary lesions in the mucosa of the mouth and nose, debilitatingly inflamed and notoriously refractory to treatment. Immunologically, this outcome has many of the same hallmarks associated with the reaction to LRV: production of type 1 interferons, bias toward a chronic Th1 inflammatory state and an impaired ability of host cells to eliminate parasites through oxidative stress. More intriguing, is that the risk of developing MCL is found almost exclusively in infections of the L. (Viannia) subtype, further indication that leishmanial metastasis is caused, at least in part, by a parasitic component. LRV present in this subgenus may contribute to the destructive inflammation of metastatic disease either by acting in concert with other intrinsic "metastatic factors" or by independently preying on host TLR3 hypersensitivity. Because LRV amplifies parasite virulence, its presence may provide a unique target for diagnostic and clinical intervention of metastatic leishmaniasis. Taking examples from other members of the Totiviridae virus family, this paper reviews the benefits and costs of endosymbiosis, specifically for the maintenance of LRV infection in Leishmania parasites, which is often at the expense of its human host.
Collapse
Affiliation(s)
- Mary-Anne Hartley
- Department of Biochemistry, University of Lausanne Epalinges, Switzerland
| | | | | | | | | |
Collapse
|
48
|
de Oliveira CI, Brodskyn CI. The immunobiology of Leishmania braziliensis infection. Front Immunol 2012; 3:145. [PMID: 22701117 PMCID: PMC3370302 DOI: 10.3389/fimmu.2012.00145] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 05/18/2012] [Indexed: 11/13/2022] Open
Abstract
Leishmaniases are a group of diseases caused by protozoa of the genus Leishmania that affect millions of people worldwide. These diseases are caused by distinct Leishmania species, of which L. braziliensis, a New World representative of the Leishmania genus, has been the least studied. Although leishmaniasis caused by L. braziliensis induces a range of clinical manifestations ranging from mild localized lesions to severe mucosal involvement, few studies have focused on elucidating the immune mechanisms behind this pathology. In this review, we focus on the immunobiology of L. braziliensis infection, emphasizing the innate and adaptive immune responses and taking into consideration both studies performed in endemic areas and experimental models of infection. Additionally, we address recent findings regarding the role of sand fly saliva in disease immunopathogenesis and vaccine development.
Collapse
|
49
|
Duell BL, Tan CK, Carey AJ, Wu F, Cripps AW, Ulett GC. Recent insights into microbial triggers of interleukin-10 production in the host and the impact on infectious disease pathogenesis. ACTA ACUST UNITED AC 2012; 64:295-313. [PMID: 22268692 DOI: 10.1111/j.1574-695x.2012.00931.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 01/17/2012] [Accepted: 01/17/2012] [Indexed: 02/06/2023]
Abstract
Since its initial description as a Th2-cytokine antagonistic to interferon-alpha and granulocyte-macrophage colony-stimulating factor, many studies have shown various anti-inflammatory actions of interleukin-10 (IL-10), and its role in infection as a key regulator of innate immunity. Studies have shown that IL-10 induced in response to microorganisms and their products plays a central role in shaping pathogenesis. IL-10 appears to function as both sword and shield in the response to varied groups of microorganisms in its capacity to mediate protective immunity against some organisms but increase susceptibility to other infections. The nature of IL-10 as a pleiotropic modulator of host responses to microorganisms is explained, in part, by its potent and varied effects on different immune effector cells which influence antimicrobial activity. A new understanding of how microorganisms trigger IL-10 responses is emerging, along with recent discoveries of how IL-10 produced during disease might be harnessed for better protective or therapeutic strategies. In this review, we summarize studies from the past 5 years that have reported the induction of IL-10 by different classes of pathogenic microorganisms, including protozoa, nematodes, fungi, viruses and bacteria and discuss the impact of this induction on the persistence and/or clearance of microorganisms in the host.
Collapse
Affiliation(s)
- Benjamin L Duell
- School of Medical Sciences, Centre for Medicine and Oral Health, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
| | | | | | | | | | | |
Collapse
|
50
|
Ronet C, Beverley SM, Fasel N. Muco-cutaneous leishmaniasis in the New World: the ultimate subversion. Virulence 2011; 2:547-52. [PMID: 21971185 DOI: 10.4161/viru.2.6.17839] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Infection by the human protozoan parasite Leishmania can lead, depending primarily on the parasite species, to either cutaneous or mucocutaneous lesions, or fatal generalized visceral infection. In the New World, Leishmania (Viannia) species can cause mucocutaneous leishmaniasis (MCL). Clinical MCL involves a strong hyper-inflammatory response and parasitic dissemination (metastasis) from a primary lesion to distant sites, leading to destructive metastatic secondary lesions especially in the nasopharyngal areas. Recently, we reported that metastasizing, but not non-metastatic strains of Leishmania (Viannia) guyanensis, have high burden of a non-segmented dsRNA virus, Leishmania RNA Virus (LRV). Viral dsRNA is sensed by the host Toll-like Receptor 3 (TLR3) thereby inducing a pro-inflammatory response and exacerbating the disease. The presence of LRV in Leishmania opens new perspectives not only in basic understanding of the intimate relation between the parasite and LRV, but also in understanding the importance of the inflammatory response in MCL patients.
Collapse
Affiliation(s)
- Catherine Ronet
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | | | | |
Collapse
|