1
|
Krayem I, Sohrabi Y, Havelková H, Gusareva ES, Strnad H, Čepičková M, Volkova V, Kurey I, Vojtíšková J, Svobodová M, Demant P, Lipoldová M. Functionally distinct regions of the locus Leishmania major response 15 control IgE or IFNγ level in addition to skin lesions. Front Immunol 2023; 14:1145269. [PMID: 37600780 PMCID: PMC10437074 DOI: 10.3389/fimmu.2023.1145269] [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: 01/15/2023] [Accepted: 05/02/2023] [Indexed: 08/22/2023] Open
Abstract
Leishmaniasis, a disease caused by parasites of Leishmania spp., endangers more than 1 billion people living in endemic countries and has three clinical forms: cutaneous, mucocutaneous, and visceral. Understanding of individual differences in susceptibility to infection and heterogeneity of its pathology is largely lacking. Different mouse strains show a broad and heterogeneous range of disease manifestations such as skin lesions, splenomegaly, hepatomegaly, and increased serum levels of immunoglobulin E and several cytokines. Genome-wide mapping of these strain differences detected more than 30 quantitative trait loci (QTLs) that control the response to Leishmania major. Some control different combinations of disease manifestations, but the nature of this heterogeneity is not yet clear. In this study, we analyzed the L. major response locus Lmr15 originally mapped in the strain CcS-9 which carries 12.5% of the genome of the resistant strain STS on the genetic background of the susceptible strain BALB/c. For this analysis, we used the advanced intercross line K3FV between the strains BALB/c and STS. We confirmed the previously detected loci Lmr15, Lmr18, Lmr24, and Lmr27 and performed genetic dissection of the effects of Lmr15 on chromosome 11. We prepared the interval-specific recombinant strains 6232HS1 and 6229FUD, carrying two STS-derived segments comprising the peak linkage of Lmr15 whose lengths were 6.32 and 17.4 Mbp, respectively, and analyzed their response to L. major infection. These experiments revealed at least two linked but functionally distinct chromosomal regions controlling IFNγ response and IgE response, respectively, in addition to the control of skin lesions. Bioinformatics and expression analysis identified the potential candidate gene Top3a. This finding further clarifies the genetic organization of factors relevant to understanding the differences in the individual risk of disease.
Collapse
Affiliation(s)
- Imtissal Krayem
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Yahya Sohrabi
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Westfälische Wilhelms-Universität, Münster, Germany
| | - Helena Havelková
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Elena S. Gusareva
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Hynek Strnad
- Department of Genomics and Bioinformatics, Institute of Molecular Genetics of The Czech Academy of Sciences, Prague, Czechia
| | - Marie Čepičková
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Valeryia Volkova
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Iryna Kurey
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Jarmila Vojtíšková
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Milena Svobodová
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
| | - Peter Demant
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Marie Lipoldová
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czechia
| |
Collapse
|
2
|
Eimanzadeh M, Mohebali M, Zarrabi M, Foroushani AR, Kazemi M, Hajjaran H, Zarei Z, Kakooei Z, Akhoundi B. The Association of Human Leucocyte Antigen (HLA) Class I and II Genes with Cutaneous and Visceral Leishmaniasis in Iranian Patients: A Preliminary Case-Control Study. IRANIAN JOURNAL OF PARASITOLOGY 2023; 18:155-164. [PMID: 37583643 PMCID: PMC10423905 DOI: 10.18502/ijpa.v18i2.13181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/19/2023] [Indexed: 08/17/2023]
Abstract
Background Leishmaniasis is currently considered a re-emerging or emerging infection based on the geographic region. The outcome of leishmaniasis vastly depends on Leishmania-host interaction. This preliminary study aimed to show the association of human leukocyte antigen (HLA) class I and II genes with healed and non-healed cutaneous leishmaniasis (CL), and symptomatic and asymptomatic visceral leishmaniasis (VL) compared with control groups in Iran. Methods Ninety-five people, including 31 patients versus 64 individuals in the control group, were enrolled. Among them, 20 patients had confirmed CL based on amastigote observation, 10 had improved CL and 10 non-healed CL. Eleven patients were suffering from confirmed VL based on direct agglutination test (Five asymptomatic and six symptomatic VL cases). Besides, they were residents in an endemic area of VL in the northwest of Iran. To select a control group, it was ensured that they had no history of leishmaniasis. Peripheral blood samples were collected from each patient. After DNA extraction, HLA typing was conducted using polymerase chain reaction - sequence-specific priming (PCR-SSP). Subsequently, data were statistically analyzed by SPSS. Results There was a statistical relationship between the presence of HLA-A26 and CL, healed CL and the existence of the B38 allele, C1 allele and symptomatic VL, as well as B1.4 allele and asymptomatic VL (P<0.05). Conclusion This primary finding indicates that several HLA genes have a potential role in the susceptibility of Iranian people to CL and VL.
Collapse
Affiliation(s)
- Mitra Eimanzadeh
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mohebali
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Center for Research of Endemic Parasites of Iran (CREPI), Tehran University of Medical Sciences, Tehran, Iran
| | | | - Abbas Rahimi Foroushani
- Department of Epidemiology and Biostatics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Homa Hajjaran
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Zabih Zarei
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Kakooei
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnaz Akhoundi
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Castellucci LC, Almeida L, Cherlin S, Fakiola M, Francis RW, Carvalho EM, Santos da Hora A, do Lago TS, Figueiredo AB, Cavalcanti CM, Alves NS, Morais KLP, Teixeira-Carvalho A, Dutra WO, Gollob KJ, Cordell HJ, Blackwell JM. A Genome-wide Association Study Identifies SERPINB10, CRLF3, STX7, LAMP3, IFNG-AS1, and KRT80 As Risk Loci Contributing to Cutaneous Leishmaniasis in Brazil. Clin Infect Dis 2021; 72:e515-e525. [PMID: 32830257 PMCID: PMC8130031 DOI: 10.1093/cid/ciaa1230] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Our goal was to identify genetic risk factors for cutaneous leishmaniasis (CL) caused by Leishmania braziliensis. METHODS Genotyping 2066 CL cases and 2046 controls using Illumina HumanCoreExomeBeadChips provided data for 4 498 586 imputed single-nucleotide variants (SNVs). A genome-wide association study (GWAS) using linear mixed models took account of genetic diversity/ethnicity/admixture. Post-GWAS positional, expression quantitative trait locus (eQTL) and chromatin interaction mapping was performed in Functional Mapping and Annotation (FUMA). Transcriptional data were compared between lesions and normal skin, and cytokines measured using flow cytometry and Bioplex assay. RESULTS Positional mapping identified 32 genomic loci associated with CL, none achieving genome-wide significance (P < 5 × 10-8). Lead SNVs at 23 loci occurred at protein coding or noncoding RNA genes, 15 with eQTLs for functionally relevant cells/tissues and/or showing differential expression in lesions. Of these, the 6 most plausible genetic risk loci were SERPINB10 (Pimputed_1000G = 2.67 × 10-6), CRLF3 (Pimputed_1000G = 5.12 × 10-6), STX7 (Pimputed_1000G = 6.06 × 10-6), KRT80 (Pimputed_1000G = 6.58 × 10-6), LAMP3 (Pimputed_1000G = 6.54 × 10-6), and IFNG-AS1 (Pimputed_1000G = 1.32 × 10-5). LAMP3 (Padjusted = 9.25 × 10-12; +6-fold), STX7 (Padjusted = 7.62 × 10-3; +1.3-fold), and CRLF3 (Padjusted = 9.19 × 10-9; +1.97-fold) were expressed more highly in CL biopsies compared to normal skin; KRT80 (Padjusted = 3.07 × 10-8; -3-fold) was lower. Multiple cis-eQTLs across SERPINB10 mapped to chromatin interaction regions of transcriptional/enhancer activity in neutrophils, monocytes, B cells, and hematopoietic stem cells. Those at IFNG-AS1 mapped to transcriptional/enhancer regions in T, natural killer, and B cells. The percentage of peripheral blood CD3+ T cells making antigen-specific interferon-γ differed significantly by IFNG-AS1 genotype. CONCLUSIONS This first GWAS for CL identified multiple genetic risk loci including a novel lead to understanding CL pathogenesis through regulation of interferon-γ by IFNG antisense RNA 1.
Collapse
Affiliation(s)
- Léa C Castellucci
- National Institute of Science and Technology in Tropical Diseases, Brazil
- Federal University of Bahia, Salvador, Brazil
| | - Lucas Almeida
- National Institute of Science and Technology in Tropical Diseases, Brazil
- Federal University of Bahia, Salvador, Brazil
| | - Svetlana Cherlin
- Population Health Sciences Institute, Newcastle University, Newcastle-Upon-Tyne, United Kingdom
| | - Michaela Fakiola
- National Institute of Molecular Genetics “Romeo ed Enrica Invernizzi,” Milan, Italy
| | - Richard W Francis
- Telethon Kids Institute, University of Western Australia, Nedlands, Australia
| | - Edgar M Carvalho
- National Institute of Science and Technology in Tropical Diseases, Brazil
| | | | | | - Amanda B Figueiredo
- International Center for Research, AC Camargo Cancer Center, São Paulo, Brazil
| | - Clara M Cavalcanti
- International Center for Research, AC Camargo Cancer Center, São Paulo, Brazil
| | - Natalia S Alves
- International Center for Research, AC Camargo Cancer Center, São Paulo, Brazil
| | - Katia L P Morais
- International Center for Research, AC Camargo Cancer Center, São Paulo, Brazil
| | | | - Walderez O Dutra
- National Institute of Science and Technology in Tropical Diseases, Brazil
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Kenneth J Gollob
- National Institute of Science and Technology in Tropical Diseases, Brazil
- International Center for Research, AC Camargo Cancer Center, São Paulo, Brazil
- Núcleo de Ensino e Pesquisa, Instituto Mario Penna, Belo Horizonte, Brazil
| | - Heather J Cordell
- Population Health Sciences Institute, Newcastle University, Newcastle-Upon-Tyne, United Kingdom
| | - Jenefer M Blackwell
- Telethon Kids Institute, University of Western Australia, Nedlands, Australia
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
4
|
Krayem I, Lipoldová M. Role of host genetics and cytokines in Leishmania infection. Cytokine 2020; 147:155244. [PMID: 33059974 DOI: 10.1016/j.cyto.2020.155244] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/20/2020] [Accepted: 08/08/2020] [Indexed: 12/29/2022]
Abstract
Cytokines and chemokines are important regulators of innate and specific responses in leishmaniasis, a disease that currently affects 12 million people. We overviewed the current information about influences of genetically engineered mouse models of cytokine and chemokine on leishmaniasis. We found that genetic background of the host, parasite species and sub-strain, as well as experimental design often modify effects of genetically engineered cytokine genes. Next we analyzed genes and QTLs (quantitative trait loci) that control response to Leishmania species in mouse in order to establish relationship between genetic control of cytokine expression and organ pathology. These studies revealed a network-like complexity of the combined effects of the multiple functionally diverse QTLs and their individual specificity. Genetic control of organ pathology and systemic immune response overlap only partially. Some QTLs control both organ pathology and systemic immune response, but the effects of genes and loci with the strongest impact on disease are cytokine-independent, whereas several loci modify cytokines levels in serum without influencing organ pathology. Understanding this genetic control might be important in development of vaccines designed to stimulate certain cytokine spectrum.
Collapse
Affiliation(s)
- Imtissal Krayem
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic
| | - Marie Lipoldová
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 14220 Prague, Czech Republic; Department of Natural Sciences, Faculty of Biomedical Engineering, Czech Technical University in Prague, Sítná 3105, 272 01 Kladno, Czech Republic.
| |
Collapse
|
5
|
da Hora AS, de Almeida LF, do Lago TS, Machado PR, Castellucci LC. FLI1 gene influences lesion size and skin test may predict therapeutic response in cutaneous leishmaniasis. Mem Inst Oswaldo Cruz 2020; 115:e190361. [PMID: 32130370 PMCID: PMC7064323 DOI: 10.1590/0074-02760190361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 01/31/2020] [Indexed: 12/18/2022] Open
Abstract
Genes associated with wound healing have been shown to be risk factors for cutaneous leishmaniasis (CL) which is caused by Leishmania braziliensis. In this study, we examined whether the genes previously associated with CL influenced the clinical outcome. Patients were genotyped and retrospectively classified as responders, who were cured with a single course of pentavalent antimony (Sbv), or as refractories, who did not respond to Sbv. Patients characterised as responders showed a stronger response to the leishmanin skin test (LST) when compared to the refractory subjects (p = 0.0003). Furthermore, we observed an association between the FLI1 CC genotype and an increased size of ulcers (p = 0.0170). We suggest that the leishmanin skin test may be a predictive tool for therapeutic outcome and reinforce FLI1 as a potential influencer of susceptibility and lesion size in CL.
Collapse
Affiliation(s)
- Anadilton Santos da Hora
- Universidade Federal da Bahia, Programa de Pós-Graduação em Ciências
da Saúde, Salvador, BA, Brasil
- Universidade Federal da Bahia, Serviço de Imunologia, Salvador, BA,
Brasil
| | | | - Tainã Souza do Lago
- Universidade Federal da Bahia, Programa de Pós-Graduação em Ciências
da Saúde, Salvador, BA, Brasil
- Universidade Federal da Bahia, Serviço de Imunologia, Salvador, BA,
Brasil
| | - Paulo Roberto Machado
- Universidade Federal da Bahia, Programa de Pós-Graduação em Ciências
da Saúde, Salvador, BA, Brasil
- Universidade Federal da Bahia, Serviço de Imunologia, Salvador, BA,
Brasil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais,
Salvador, BA, Brasil
| | - Léa Cristina Castellucci
- Universidade Federal da Bahia, Programa de Pós-Graduação em Ciências
da Saúde, Salvador, BA, Brasil
- Universidade Federal da Bahia, Serviço de Imunologia, Salvador, BA,
Brasil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais,
Salvador, BA, Brasil
| |
Collapse
|
6
|
Lopes DM, Oliveira SC, Page B, Carvalho LP, Carvalho EM, Cardoso LS. Schistosoma mansoni rSm29 Antigen Induces a Regulatory Phenotype on Dendritic Cells and Lymphocytes From Patients With Cutaneous Leishmaniasis. Front Immunol 2019; 9:3122. [PMID: 30687325 PMCID: PMC6333737 DOI: 10.3389/fimmu.2018.03122] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 12/18/2018] [Indexed: 12/24/2022] Open
Abstract
The immune response induced by Schistosma mansoni antigens is able to prevent immune-mediated diseases. Conversely, the inflammatory response in cutaneous leishmaniasis (CL), although responsible for controlling the infection, is also associated with the pathogenesis of disease. The aim of this study was to evaluate the potential of the S. mansoni Sm29 antigen to change certain aspects of the profiles of monocyte derived dendritic cells (MoDCs) and lymphocytes from subjects with CL in vitro. Expression of surface molecules and intracellular cytokines in the MoDCs and lymphocytes as well as the proliferation of Leishmania braziliensis were evaluated by flow cytometry. Levels of cytokines were evaluated in culture supernatants by ELISA. It was observed that stimulation by rSm29 increased the frequency of expression of CD83, CD80, CD86, and IL-10R in MoDCs compared to non-stimulated cultures. Additionally rSm29 decreased the frequency CD4+ and CD8+ T cells expressing CD28 and increased the frequency of CD4+CD25hi and CD4+CTLA-4+ T lymphocytes. Addition of rSm29 to cultures increased IL-10 levels and decreased levels of IL-12p40 and IFN-γ, while not altering TNF levels compared to non-stimulated cultures. This study showed that rSm29 induced a regulatory profile in MoDCs and lymphocytes and thereby regulated the exaggerated inflammation observed in CL. Considering that there are few therapeutic options for leishmaniasis, the use of rSm29 may be an alternative to current treatment and may be an important strategy to reduce the healing time of lesions in patients with CL.
Collapse
Affiliation(s)
- Diego Mota Lopes
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil.,Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (DT/CNPq), Brasilia, Brazil
| | - Sérgio Costa Oliveira
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (DT/CNPq), Brasilia, Brazil.,Departamento de Bioquímica e Imunologia, Instituto de ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Brady Page
- Massachusetts General Hospital, Boston, MA, United States
| | - Lucas P Carvalho
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil.,Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (DT/CNPq), Brasilia, Brazil.,Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Edgar M Carvalho
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil.,Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (DT/CNPq), Brasilia, Brazil.,Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Luciana Santos Cardoso
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil.,Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (DT/CNPq), Brasilia, Brazil.,Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, UFBA, Salvador, Brazil
| |
Collapse
|
7
|
Autoimmune-Disease-Prone NOD Mice Help To Reveal a New Genetic Locus for Reducing Pulmonary Disease Caused by Mycoplasma pulmonis. Infect Immun 2018; 86:IAI.00812-17. [PMID: 29263105 DOI: 10.1128/iai.00812-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 12/07/2017] [Indexed: 11/20/2022] Open
Abstract
Mycoplasmas are bacterial pathogens of a range of animals, including humans, and are a common cause of respiratory disease. However, the host genetic factors that affect resistance to infection or regulate the resulting pulmonary inflammation are not well defined. We and others have previously demonstrated that nonobese diabetic (NOD) mice can be used to investigate disease loci that affect bacterial infection and autoimmune diabetes. Here we show that NOD mice are more susceptible than C57BL/6 (B6) mice to infection with Mycoplasma pulmonis, a natural model of pulmonary mycoplasmosis. The lungs of infected NOD mice had higher loads of M. pulmonis and more severe inflammatory lesions. Moreover, congenic NOD mice that harbored different B6-derived chromosomal intervals enabled identification and localization of a new mycoplasmosis locus, termed Mpr2, on chromosome 13. These congenic NOD mice demonstrated that the B6 allele for Mpr2 reduced the severity of pulmonary inflammation caused by infection with M. pulmonis and that this was associated with altered cytokine and chemokine concentrations in the infected lungs. Mpr2 also colocalizes to the same genomic interval as Listr2 and Idd14, genetic loci linked to listeriosis resistance and autoimmune diabetes susceptibility, respectively, suggesting that allelic variation within these loci may affect the development of both infectious and autoimmune disease.
Collapse
|
8
|
de Vasconcelos TCB, Furtado MC, Belo VS, Morgado FN, Figueiredo FB. Canine susceptibility to visceral leishmaniasis: A systematic review upon genetic aspects, considering breed factors and immunological concepts. INFECTION GENETICS AND EVOLUTION 2017; 74:103293. [PMID: 28987807 DOI: 10.1016/j.meegid.2017.10.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/30/2017] [Accepted: 10/03/2017] [Indexed: 01/01/2023]
Abstract
Dogs have different susceptibility degrees to leishmaniasis; however, genetic research on this theme is scarce, manly on visceral form. The aims of this systematic review were to describe and discuss the existing scientific findings on genetic susceptibility to canine leishmaniasis, as well as to show the gaps of the existing knowledge. Twelve articles were selected, including breed immunological studies, genome wide associations or other gene polymorphism or gene sequencing studies, and transcription approaches. As main results of literature, there was a suggestion of genetic clinical resistance background for Ibizan Hound dogs, and alleles associated with protection or susceptibility to visceral leishmaniasis in Boxer dogs. Genetic markers can explain phenotypic variance in both pro- and anti-inflammatory cytokines and in cellular immune responses, including antigen presentation. Many gene segments are involved in canine visceral leishmaniasis phenotype, with Natural Resistance Associated Macrophage Protein 1 (NRAMP1) as the most studied. This was related to both protection and susceptibility. In comparison with murine and human genetic approaches, lack of knowledge in dogs is notorious, with many possibilities for new studies, revealing a wide field to be assessed on canine leishmaniasis susceptibility research.
Collapse
Affiliation(s)
- Tassia Cristina Bello de Vasconcelos
- Centro de Controle de Zoonoses, Vigilância em Saúde, Secretaria Municipal de Saúde, Prefeitura Municipal de Resende, Rua Euridices Paulina de Almeida, 300, Vicentina II, Resende, RJ 27500-000, Brazil.
| | - Marina Carvalho Furtado
- Fiocruz Mata Atlântica, Fundação Oswaldo Cruz, Estrada Rodrigues Caldas, 3400, Taquara, Rio de Janeiro, RJ 22713-375, Brazil
| | - Vinícius Silva Belo
- Universidade Federal de São João del-Rei, campus Centro Oeste Dona Lindu, Rua Sebastião Gonçalves Coelho, 400, Chanadour, Divinópolis, MG 35.501-296, Brazil
| | - Fernanda Nazaré Morgado
- Laboratório de Pesquisa em Leishmaniose, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil, 4365, Manguinhos, Rio de Janeiro, RJ 21040-360, Brazil
| | - Fabiano Borges Figueiredo
- Laboratório de Biologia Celular, Instituto Carlos Chagas, Rua Professor Algacyr Munhoz Mader, 3.775, CIC, campus do Tecpar, bloco C, Curitiba, PR 81.350-010 Brazil
| |
Collapse
|
9
|
The role of monocytes in models of infection by protozoan parasites. Mol Immunol 2017; 88:174-184. [PMID: 28704704 DOI: 10.1016/j.molimm.2017.06.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 05/29/2017] [Accepted: 06/04/2017] [Indexed: 02/07/2023]
Abstract
The confirmation of developmental differences between tissue macrophages and peripheral monocytes has changed our view of the functions and dynamics of these two important components of the innate immune system. It has been demonstrated conclusively that homeostasis of tissue resident macrophages is maintained by a low proliferative turn over. During an inflammatory response, bone marrow derived monocytes enter the tissue in large numbers and take part in the defense against the pathogens. After the destruction of invading pathogens, these cells disappear and tissue resident macrophages can be detected again. This new appreciation of the innate immune response has not only answered many outstanding questions regarding the role of the different myeloid cell types in inflammation, but also opened up new areas of research relating to the tissue- and pathogen-specific fate of the inflammatory macrophages or dendritic cells (DCs), and the transfer of this knowledge from mouse models to the human immune system. Nevertheless, there is still confusion in infection models, and especially in studies of human infections, as to what extent these recent observations and findings influence previous interpretations of data. This review will focus on insights from mouse models, summarize the literature on the ontogeny of macrophages and monocytes, explain the role of frequently used monocyte markers and effector molecules, and finally, discuss the role of inflammatory monocytes/macrophages/DCs in two experimental parasitic diseases.
Collapse
|
10
|
Almeida L, Silva JA, Andrade VM, Machado P, Jamieson SE, Carvalho EM, Blackwell JM, Castellucci LC. Analysis of expression of FLI1 and MMP1 in American cutaneous leishmaniasis caused by Leishmania braziliensis infection. INFECTION GENETICS AND EVOLUTION 2017; 49:212-220. [PMID: 28119029 DOI: 10.1016/j.meegid.2017.01.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 01/10/2017] [Accepted: 01/18/2017] [Indexed: 12/19/2022]
Abstract
FLI1 (Friend leukemia virus integration 1) and IL6 (interleukin 6; IL-6) are associated with Leishmania braziliensis susceptibility. Cutaneous lesions show exaggerated matrix metalloproteinase 1 (MMP1). In other skin diseases, FLI1 promoter methylation reduces FLI1 expression, and low FLI1 down-regulates MMP1. IL-6 increases FLI1 expression. We hypothesized that epigenetic regulation of FLI1 in cutaneous leishmaniasis, together with IL-6, might determine MMP1 expression. While generally low (<10%), percent FLI1 promoter methylation was lower (P=0.001) in lesion biopsies than normal skin. Contrary to expectation, a strong positive correlation occurred between FLI1 methylation and gene expression in lesions (r=0.98, P=0.0005) and in IL-6-treated L. braziliensis-infected macrophages (r=0.99, P=0.0004). In silico analysis of the FLI1 promoter revealed co-occurring active H3K27ac and repressive DNA methylation marks to enhance gene expression. FLI1 expression was enhanced between 3 and 24hour post infection in untreated (P=0.0002) and IL-6-treated (P=0.028) macrophages. MMP1 was enhanced in lesion biopsies (P=0.0002), induced (P=0.007) in infected macrophages, but strongly inhibited by IL-6. No correlations occurred between FLI1 and MMP1 expression in lesions or infected macrophages (with/without IL-6). We conclude that MMP1 is regulated by factors other than FLI1, and that the influence of IL-6 on MMP1 was independent of its effect on FLI1.
Collapse
Affiliation(s)
- Lucas Almeida
- National Institute of Cience and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil.,Program of Post-graduation in Health Sciences, Federal University of Bahia, Salvador, Brazil
| | - Juliana A Silva
- National Institute of Cience and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil.,Program of Post-graduation in Health Sciences, Federal University of Bahia, Salvador, Brazil
| | - Viviane M Andrade
- National Institute of Cience and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil.,Program of Post-graduation in Health Sciences, Federal University of Bahia, Salvador, Brazil
| | - Paulo Machado
- National Institute of Cience and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil
| | - Sarra E Jamieson
- Telethon Kids Institute, The University of Western Australia, Subiaco, Western Australia, Australia
| | - Edgar M Carvalho
- National Institute of Cience and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil
| | - Jenefer M Blackwell
- Telethon Kids Institute, The University of Western Australia, Subiaco, Western Australia, Australia.,Department of Pathology and Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Léa C Castellucci
- National Institute of Cience and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil
| |
Collapse
|
11
|
Fromm PD, Kling JC, Remke A, Bogdan C, Körner H. Fatal Leishmaniasis in the Absence of TNF Despite a Strong Th1 Response. Front Microbiol 2016; 6:1520. [PMID: 26834705 PMCID: PMC4722107 DOI: 10.3389/fmicb.2015.01520] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/17/2015] [Indexed: 01/28/2023] Open
Abstract
Induction of inducible nitric oxide synthase in mononuclear phagocytes by IFN-γ and innate tumor necrosis factor (TNF) provide the basis for an effective immune response to the intracellular parasite Leishmania (L.) major. In previous experiments, we observed a fatal visceral form of leishmaniasis in L. major-infected C57BL/6 TNF-/- mice. To further delineate the protective function of TNF and its receptor requirements, we comparatively assessed L. major-infected C57BL/6 mice that were either deficient for membrane and soluble TNF (Tnf-/-), for soluble TNF alone (memTnfΔ/Δ), or the TNF receptors type 1 (Tnfr1-/-) or type 2 (Tnfr2-/-). We detected locally and systemically increased levels of the cytokine IFN-γ in the absence of the TNF-TNFR1-signaling pathway. An analysis of transcription factors and cytokines revealed that activated Tnf-/- CD4+ T cells displayed a highly active Th1 phenotype with a strong usage of the T cell receptor Vβ5.1/2. From these data we conclude that the fatal outcome of L. major infection in Tnf-/- mice does not result from a skewed or deficient Th1 differentiation.
Collapse
Affiliation(s)
- Phillip D Fromm
- Comparative Genomics Centre, James Cook University, Townsville QN, Australia
| | - Jessica C Kling
- Menzies Institute for Medical Research Tasmania, HobartTAS, Australia; Blumenthal Group, The University of Queensland Diamantina Institute, Translational Research Institute, WoolloongabbaQLD, Australia
| | - Annika Remke
- Menzies Institute for Medical Research Tasmania, Hobart TAS, Australia
| | - Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Friederich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen Erlangen, Germany
| | - Heinrich Körner
- Menzies Institute for Medical Research Tasmania, Hobart TAS, Australia
| |
Collapse
|
12
|
Almeida L, Oliveira J, Guimarães LH, Carvalho EM, Blackwell JM, Castellucci L. Wound healing genes and susceptibility to cutaneous leishmaniasis in Brazil: role of COL1A1. INFECTION GENETICS AND EVOLUTION 2015; 30:225-229. [PMID: 25562121 DOI: 10.1016/j.meegid.2014.12.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 12/22/2014] [Accepted: 12/27/2014] [Indexed: 02/06/2023]
Abstract
Previous studies have demonstrated a role for wound healing genes in resolution of cutaneous lesions caused by Leishmania spp. in both mice and humans, including the gene FLI1 encoding Friend leukemia virus integration 1. Reduction of Fli1 expression in mice has been shown to result in up-regulation of collagen type I alpha 1 (Col1a1) and alpha 2 (Col1a2) genes and, conversely, in down-regulation of the matrix metalloproteinase 1 (Mmp1) gene, suggesting that Fli1 suppression is involved in activation of the profibrotic gene program. Here we examined single nucleotide polymorphisms (SNPs) in these genes as risk factors for cutaneous (CL) and mucosal leishmaniasis (ML), and leishmaniasis per se, caused by L. braziliensis in humans. SNPs were genotyped in 168 nuclear families (250 CL; 87 ML cases) and replicated in 157 families (402 CL; 39 ML cases). Family-based association tests (FBAT) showed the strongest association between SNPs rs1061237 (combined P=0.002) and rs2586488 (combined P=0.027) at COL1A1 and CL disease. This contributes to our further understanding of the role of wound healing in the resolution of CL disease, providing potential for therapies modulating COL1A1 via drugs acting on FLI1.
Collapse
Affiliation(s)
- Lucas Almeida
- National Institute of Science and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil.
| | - Joyce Oliveira
- National Institute of Science and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil.
| | - Luiz Henrique Guimarães
- National Institute of Science and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil.
| | - Edgar M Carvalho
- National Institute of Science and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil.
| | - Jenefer M Blackwell
- Telethon Kids Institute, The University of Western Australia, Subiaco, Western Australia, Australia; Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK.
| | - Léa Castellucci
- National Institute of Science and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil.
| |
Collapse
|
13
|
Raza S, Barnett MW, Barnett-Itzhaki Z, Amit I, Hume DA, Freeman TC. Analysis of the transcriptional networks underpinning the activation of murine macrophages by inflammatory mediators. J Leukoc Biol 2014; 96:167-83. [PMID: 24721704 PMCID: PMC4378362 DOI: 10.1189/jlb.6hi0313-169r] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 01/17/2014] [Accepted: 02/23/2014] [Indexed: 01/09/2023] Open
Abstract
Macrophages respond to the TLR4 agonist LPS with a sequential transcriptional cascade controlled by a complex regulatory network of signaling pathways and transcription factors. At least two distinct pathways are currently known to be engaged by TLR4 and are distinguished by their dependence on the adaptor molecule MyD88. We have used gene expression microarrays to define the effects of each of three variables--LPS dose, LPS versus IFN-β and -γ, and genetic background--on the transcriptional response of mouse BMDMs. Analysis of correlation networks generated from the data has identified subnetworks or modules within the macrophage transcriptional network that are activated selectively by these variables. We have identified mouse strain-specific signatures, including a module enriched for SLE susceptibility candidates. In the modules of genes unique to different treatments, we found a module of genes induced by type-I IFN but not by LPS treatment, suggesting another layer of complexity in the LPS-TLR4 signaling feedback control. We also observe that the activation of the complement system, in common with the known activation of MHC class 2 genes, is reliant on IFN-γ signaling. Taken together, these data further highlight the exquisite nature of the regulatory systems that control macrophage activation, their likely relevance to disease resistance/susceptibility, and the appropriate response of these cells to proinflammatory stimuli.
Collapse
Affiliation(s)
- Sobia Raza
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Scotland, United Kingdom; and
| | - Mark W Barnett
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Scotland, United Kingdom; and
| | | | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - David A Hume
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Scotland, United Kingdom; and
| | - Tom C Freeman
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Scotland, United Kingdom; and
| |
Collapse
|
14
|
The ets transcription factor Fli-1 in development, cancer and disease. Oncogene 2014; 34:2022-31. [PMID: 24909161 PMCID: PMC5028196 DOI: 10.1038/onc.2014.162] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/03/2014] [Accepted: 05/04/2014] [Indexed: 12/13/2022]
Abstract
Friend Leukemia Virus Induced erythroleukemia-1 (Fli-1), an ETS transcription factor, was isolated a quarter century ago through a retrovirus mutagenesis screen. Fli-1 has since been recognized to play critical roles in normal development and homeostasis. For example, it transcriptionally regulates genes that drive normal hematopoiesis and vasculogenesis. Indeed, Fli-1 is one of 10 key regulators of hematopoietic stem/progenitor cell maintenance and differentiation. Aberrant expression of Fli-1 also underlies a number of virally induced leukemias, including Friend virus-induced erythroleukemia and various types of human cancers, and it is the target of chromosomal translocations in childhood Ewing’s sarcoma. Abnormal expression of Fli-1 is important in the aetiology of auto-immune diseases such as Systemic Lupus Erythematosus (SLE) and Systemic Sclerosis (SSc). These studies establish Fli-1 as a strong candidate for drug development. Despite difficulties in targeting transcription factors, recent studies identified small molecule inhibitors for Fli-1. Here we review past and ongoing research on Fli-1 with emphasis on its mechanistic function in autoimmune disease and malignant transformation. The significance of identifying Fli-1 inhibitors and their clinical applications for treatment of disease and cancer with deregulated Fli-1 expression are discussed.
Collapse
|
15
|
Castellucci LC, Almeida LFD, Jamieson SE, Fakiola M, Carvalho EMD, Blackwell JM. Host genetic factors in American cutaneous leishmaniasis: a critical appraisal of studies conducted in an endemic area of Brazil. Mem Inst Oswaldo Cruz 2014; 109:279-88. [PMID: 24863979 PMCID: PMC4131779 DOI: 10.1590/0074-0276140028] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 04/07/2014] [Indexed: 12/17/2022] Open
Abstract
American cutaneous leishmaniasis (ACL) is a vector-transmitted infectious disease with an estimated 1.5 million new cases per year. In Brazil, ACL represents a significant public health problem, with approximately 30,000 new reported cases annually, representing an incidence of 18.5 cases per 100,000 inhabitants. Corte de Pedra is in a region endemic for ACL in the state of Bahia (BA), northeastern Brazil, with 500-1,300 patients treated annually. Over the last decade, population and family-based candidate gene studies were conducted in Corte de Pedra, founded on previous knowledge from studies on mice and humans. Notwithstanding limitations related to sample size and power, these studies contribute important genetic biomarkers that identify novel pathways of disease pathogenesis and possible new therapeutic targets. The present paper is a narrative review about ACL immunogenetics in BA, highlighting in particular the interacting roles of the wound healing gene FLI1 with interleukin-6 and genes SMAD2 and SMAD3 of the transforming growth factor beta signalling pathway. This research highlights the need for well-powered genetic and functional studies on Leishmania braziliensis infection as essential to define and validate the role of host genes in determining resistance/susceptibility regarding this disease.
Collapse
Affiliation(s)
- Léa Cristina Castellucci
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Universidade Federal da Bahia, Salvador, BA, Brasil
| | - Lucas Frederico de Almeida
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Universidade Federal da Bahia, Salvador, BA, Brasil
| | | | - Michaela Fakiola
- Telethon Kids Institute, The University of Western Australi, Perth, Australia
| | - Edgar Marcelino de Carvalho
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Universidade Federal da Bahia, Salvador, BA, Brasil
| | | |
Collapse
|
16
|
Bakhshi H, Oshaghi MA, Abai MR, Rassi Y, Akhavan AA, Mohebali M, Hajaran H, Mohtarami F, Mirzajani H, Maleki-Ravasan N. MtDNA CytB Structure of Rhombomys opimus (Rodentia: Gerbellidae), the Main Reservoir of Cutaneous Leishmaniasis in the Borderline of Iran-Turkmenistan. J Arthropod Borne Dis 2013; 7:173-84. [PMID: 24409443 PMCID: PMC3875884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 08/18/2013] [Indexed: 10/25/2022] Open
Abstract
BACKGROUND Great gerbils, Rhombomys opimus, are the main reservoir host of zoonootic cutaneous leishmaniasis (ZCL) in Iran and neighboring countries. Based on morphological traits two subspecies R. opimus sodalis and R. opimus sargadensis have reported in the country. However, variation in infection rate and signs to Leishmania parasites, phenotype, size, and sexual polymorphisms demand more details to elucidate clearly the role of great gerbils in ZCL epidemiology. METHODS PCR-RFLP and PCR-direct sequencing were used to analyze mitochondrial DNA cytochrome B (mtDNA-cytB) gene structure of R. opimus collected from Golestan and Khorasan-e-Razavi Provinces in 2011 that are neighbor to Turkmenistan Country where ZCL is endemic in both sides of the borderline. RESULTS All of the specimens (n= 61) were morphologically or genetically similar to the typical R. opimus sodalis. However, there were 9 (1.5%) DNA substitutions throughout the 583 bp of the Cyt b gene of the samples sequenced comprising six DNA haplotypes. Maximum likelihood or neighbor joining phylogenetic trees inferred from the sequences could resolve the populations according to their subspecies as well as geographical origins. DISCUSSION The DNA polymorphisms in the great gerbils may correspond to the signs and infection rate in the animal. However, further studies are needed to match these six haplotypes with different signs and parasite sustaining following infection with L. major in the great gerbils.
Collapse
Affiliation(s)
- Hasan Bakhshi
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Oshaghi
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran,Corresponding author: Dr Mohammad Ali Oshaghi, E-mail:
| | - Mohammad Reza Abai
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Yavar Rassi
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ahmad Akhavan
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mohebali
- Department of Medical Parasitology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Homa Hajaran
- Department of Medical Parasitology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mohtarami
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Naseh Maleki-Ravasan
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Sohrabi Y, Havelková H, Kobets T, Šíma M, Volkova V, Grekov I, Jarošíková T, Kurey I, Vojtíšková J, Svobodová M, Demant P, Lipoldová M. Mapping the genes for susceptibility and response to Leishmania tropica in mouse. PLoS Negl Trop Dis 2013; 7:e2282. [PMID: 23875032 PMCID: PMC3708836 DOI: 10.1371/journal.pntd.0002282] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 05/09/2013] [Indexed: 12/04/2022] Open
Abstract
Background L. tropica can cause both cutaneous and visceral leishmaniasis in humans. Although the L. tropica-induced cutaneous disease has been long known, its potential to visceralize in humans was recognized only recently. As nothing is known about the genetics of host responses to this infection and their clinical impact, we developed an informative animal model. We described previously that the recombinant congenic strain CcS-16 carrying 12.5% genes from the resistant parental strain STS/A and 87.5% genes from the susceptible strain BALB/c is more susceptible to L. tropica than BALB/c. We used these strains to map and functionally characterize the gene-loci regulating the immune responses and pathology. Methods We analyzed genetics of response to L. tropica in infected F2 hybrids between BALB/c×CcS-16. CcS-16 strain carries STS-derived segments on nine chromosomes. We genotyped these segments in the F2 hybrid mice and tested their linkage with pathological changes and systemic immune responses. Principal Findings We mapped 8 Ltr (Leishmania tropica response) loci. Four loci (Ltr2, Ltr3, Ltr6 and Ltr8) exhibit independent responses to L. tropica, while Ltr1, Ltr4, Ltr5 and Ltr7 were detected only in gene-gene interactions with other Ltr loci. Ltr3 exhibits the recently discovered phenomenon of transgenerational parental effect on parasite numbers in spleen. The most precise mapping (4.07 Mb) was achieved for Ltr1 (chr.2), which controls parasite numbers in lymph nodes. Five Ltr loci co-localize with loci controlling susceptibility to L. major, three are likely L. tropica specific. Individual Ltr loci affect different subsets of responses, exhibit organ specific effects and a separate control of parasite load and organ pathology. Conclusion We present the first identification of genetic loci controlling susceptibility to L. tropica. The different combinations of alleles controlling various symptoms of the disease likely co-determine different manifestations of disease induced by the same pathogen in individual mice. Leishmaniasis, a disease caused by Leishmania ssp. is among the most neglected infectious diseases. In humans, L. tropica causes cutaneous form of leishmaniasis, but can damage internal organs too. The reasons for this variability are not known, and its genetic basis was never investigated. Therefore, analysis of genes affecting host's responses to this infection can elucidate the characteristics of individual host-parasite interactions. Recombinant congenic strain CcS-16 carries 12.5% genes from the mouse strain STS/A on genetic background of the strain BALB/c, and it is more susceptible than BALB/c. In F2 hybrids between BALB/c and CcS-16 we detected and mapped eight gene-loci, Ltr1-8 (Leishmania tropica response 1-8) that control various manifestations of disease: skin lesions, splenomegaly, hepatomegaly, parasite numbers in spleen, liver, and inguinal lymph nodes, and serum level of CCL3, CCL5, and CCL7 after L. tropica infection. These loci are functionally heterogeneous - each influences a different set of responses to the pathogen. Five loci co-localize with the previously described loci that control susceptibility to L. major, three are species-specific. Ltr2 co-localizes not only with Lmr14 (Leishmania major response 14), but also with Ir2 influencing susceptibility to L. donovani and might therefore carry a common gene controlling susceptibility to leishmaniasis.
Collapse
Affiliation(s)
- Yahya Sohrabi
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Helena Havelková
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Tetyana Kobets
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Matyáš Šíma
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Valeriya Volkova
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Igor Grekov
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Taťána Jarošíková
- Faculty of Biomedical Engineering, Czech Technical University in Prague, Kladno, Czech Republic
| | - Iryna Kurey
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Jarmila Vojtíšková
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | - Peter Demant
- Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Marie Lipoldová
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- * E-mail:
| |
Collapse
|
18
|
Costa ISF, de Souza GFP, de Oliveira MG, Abrahamsohn IDA. S-nitrosoglutathione (GSNO) is cytotoxic to intracellular amastigotes and promotes healing of topically treated Leishmania major or Leishmania braziliensis skin lesions. J Antimicrob Chemother 2013; 68:2561-8. [DOI: 10.1093/jac/dkt210] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
19
|
Nylén S, Eidsmo L. Tissue damage and immunity in cutaneous leishmaniasis. Parasite Immunol 2012; 34:551-61. [DOI: 10.1111/pim.12007] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 09/17/2012] [Indexed: 12/21/2022]
Affiliation(s)
- S. Nylén
- Department of Microbiology; Tumor and Cell Biology; Karolinska Institutet; Stockholm; Sweden
| | - L. Eidsmo
- Molecular Dermatology; Department of Medicine Solna; Karolinska Institutet; Stockholm; Sweden
| |
Collapse
|
20
|
Bujor AM, Haines P, Padilla C, Christmann RB, Junie M, Sampaio-Barros PD, Lafyatis R, Trojanowska M. Ciprofloxacin has antifibrotic effects in scleroderma fibroblasts via downregulation of Dnmt1 and upregulation of Fli1. Int J Mol Med 2012; 30:1473-80. [PMID: 23041765 PMCID: PMC4035782 DOI: 10.3892/ijmm.2012.1150] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 08/30/2012] [Indexed: 12/14/2022] Open
Abstract
Systemic sclerosis (SSc) is characterized by fibrosis of the skin and internal organs. The present study was undertaken to examine the effects of ciprofloxacin, a fluoroquinolone antibiotic implicated in matrix remodeling, on dermal and lung fibroblasts obtained from SSc patients. Dermal and lung fibroblasts from SSc patients and healthy subjects were treated with ciprofloxacin. Western blotting was used to analyze protein levels and RT-PCR was used to measure mRNA expression. The pharmacologic inhibitor UO126 was used to block Erk1/2 signaling. SSc dermal fibroblasts demonstrated a significant decrease in collagen type I mRNA and protein levels after antibiotic treatment, while healthy dermal fibroblasts were less sensitive to ciprofloxacin, downregulating collagen only at the protein levels. Connective tissue growth factor (CCN2) gene expression was significantly reduced and matrix metalloproteinase 1 (MMP1) levels were enhanced after ciprofloxacin treatment to a similar extent in healthy and SSc fibroblasts. Ciprofloxacin induced Erk1/2 phosphorylation, and Erk1/2 blockade completely prevented MMP1 upregulation. However, Smad1 and Smad3 activation in response to TGFβ was not affected. The expression of friend leukemia integration factor 1 (Fli1), a transcriptional repressor of collagen, was increased after treatment with ciprofloxacin only in SSc fibroblasts, and this was accompanied by a decrease in the levels of DNA methyltransferase 1 (Dnmt1). Similar effects were observed in SSc-interstitial lung disease (ILD) lung fibroblasts. In summary, our study demonstrates that ciprofloxacin has antifibrotic actions in SSc dermal and lung fibroblasts via the downregulation of Dnmt1, the upregulation of Fli1 and induction of MMP1 gene expression via an Erk1/2-dependent mechanism. Thus, our data suggest that ciprofloxacin may be an attractive therapy for SSc skin and lung fibrosis.
Collapse
Affiliation(s)
- Andreea M Bujor
- Arthritis Center, Rheumatology, Boston University School of Medicine, Boston, MA 02118, USA.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Castellucci L, Jamieson SE, Almeida L, Oliveira J, Guimarães LH, Lessa M, Fakiola M, de Jesus AR, Miller EN, Carvalho EM, Blackwell JM. Wound healing genes and susceptibility to cutaneous leishmaniasis in Brazil. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2012; 12:1102-10. [PMID: 22554650 PMCID: PMC3372530 DOI: 10.1016/j.meegid.2012.03.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 03/15/2012] [Accepted: 03/17/2012] [Indexed: 12/16/2022]
Abstract
Leishmania braziliensis causes cutaneous (CL) and mucosal (ML) leishmaniasis. In the mouse, Fli1 was identified as a gene influencing enhanced wound healing and resistance to CL caused by Leishmania major. Polymorphism at FLI1 is associated with CL caused by L. braziliensis in humans, with an inverse association observed for ML disease. Here we extend the analysis to look at other wound healing genes, including CTGF, TGFB1, TGFBR1/2, SMADS 2/3/4/7 and FLII, all functionally linked along with FLI1 in the TGF beta pathway. Haplotype tagging single nucleotide polymorphisms (tag-SNPs) were genotyped using Taqman technology in 325 nuclear families (652 CL cases; 126 ML cases) from Brazil. Robust case-pseudocontrol (CPC) conditional logistic regression analysis showed associations between CL and SNPs at CTGF (SNP rs6918698; CC genotype; OR 1.67; 95%CI 1.10-2.54; P=0.016), TGFBR2 (rs1962859; OR 1.50; 95%CI 1.12-1.99; P=0.005), SMAD2 (rs1792658; OR 1.57; 95%CI 1.04-2.38; P=0.03), SMAD7 (rs4464148; AA genotype; OR 2.80; 95%CI 1.00-7.87; P=0.05) and FLII (rs2071242; OR 1.60; 95%CI 1.14-2.24; P=0.005), and between ML and SNPs at SMAD3 (rs1465841; OR 2.15; 95%CI 1.13-4.07; P=0.018) and SMAD7 (rs2337107; TT genotype; OR 3.70; 95%CI 1.27-10.7; P=0.016). Stepwise logistic regression analysis showed that all SNPs associated with CL at FLI1, CTGF, TGFBR2, and FLII showed independent effects from each other, but SNPs at SMAD2 and SMAD7 did not add independent effects to SNPs from other genes. These results suggest that TGFβ signalling via SMAD2 is important in directing events that contribute to CL, whereas signalling via SMAD3 is important in ML. Both are modulated by the inhibitory SMAD7 that acts upstream of SMAD2 and SMAD3 in this signalling pathway. Along with the published FLI1 association, these data further contribute to the hypothesis that wound healing processes are important determinants of pathology associated with cutaneous forms of leishmaniasis.
Collapse
Affiliation(s)
- Léa Castellucci
- National Institute of Cience and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil
| | - Sarra E Jamieson
- Telethon Institute for Child Health Research, Centre for Child Health Research, The University of Western Australia, Subiaco, Western Australia, Australia
| | - Lucas Almeida
- National Institute of Cience and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil
| | - Joyce Oliveira
- National Institute of Cience and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil
| | - Luiz Henrique Guimarães
- National Institute of Cience and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil
| | - Marcus Lessa
- National Institute of Cience and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil
| | - Michaela Fakiola
- Telethon Institute for Child Health Research, Centre for Child Health Research, The University of Western Australia, Subiaco, Western Australia, Australia
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Amélia Ribeiro de Jesus
- National Institute of Cience and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil
- Instituto de Investigação em Imunologia, São Paulo, Brazil
- Universidade Federal de Sergipe – Aracaju, Brazil
| | - E. Nancy Miller
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Edgar M Carvalho
- National Institute of Cience and Technology in Tropical Diseases, Brazil and Federal University of Bahia, Salvador, Brazil
| | - Jenefer M Blackwell
- Telethon Institute for Child Health Research, Centre for Child Health Research, The University of Western Australia, Subiaco, Western Australia, Australia
- Cambridge Institute for Medical Research and Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| |
Collapse
|
22
|
Genetics of host response to Leishmania tropica in mice - different control of skin pathology, chemokine reaction, and invasion into spleen and liver. PLoS Negl Trop Dis 2012; 6:e1667. [PMID: 22679519 PMCID: PMC3367980 DOI: 10.1371/journal.pntd.0001667] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 04/17/2012] [Indexed: 01/12/2023] Open
Abstract
Background Leishmaniasis is a disease caused by protozoan parasites of genus Leishmania. The frequent involvement of Leishmania tropica in human leishmaniasis has been recognized only recently. Similarly as L. major, L. tropica causes cutaneous leishmaniasis in humans, but can also visceralize and cause systemic illness. The relationship between the host genotype and disease manifestations is poorly understood because there were no suitable animal models. Methods We studied susceptibility to L. tropica, using BALB/c-c-STS/A (CcS/Dem) recombinant congenic (RC) strains, which differ greatly in susceptibility to L. major. Mice were infected with L. tropica and skin lesions, cytokine and chemokine levels in serum, and parasite numbers in organs were measured. Principal Findings Females of BALB/c and several RC strains developed skin lesions. In some strains parasites visceralized and were detected in spleen and liver. Importantly, the strain distribution pattern of symptoms caused by L. tropica was different from that observed after L. major infection. Moreover, sex differently influenced infection with L. tropica and L. major. L. major-infected males exhibited either higher or similar skin pathology as females, whereas L. tropica-infected females were more susceptible than males. The majority of L. tropica-infected strains exhibited increased levels of chemokines CCL2, CCL3 and CCL5. CcS-16 females, which developed the largest lesions, exhibited a unique systemic chemokine reaction, characterized by additional transient early peaks of CCL3 and CCL5, which were not present in CcS-16 males nor in any other strain. Conclusion Comparison of L. tropica and L. major infections indicates that the strain patterns of response are species-specific, with different sex effects and largely different host susceptibility genes. Several hundred million people are exposed to the risk of leishmaniasis, a disease caused by intracellular protozoan parasites of several Leishmania species and transmitted by phlebotomine sand flies. In humans, L. tropica causes cutaneous form of leishmaniasis with painful and long-persisting lesions in the site of the insect bite, but the parasites can also penetrate to internal organs. The relationship between the host genes and development of the disease was demonstrated for numerous infectious diseases. However, the search for susceptibility genes in the human population could be a difficult task. In such cases, animal models may help to discover the role of different genes in interactions between the parasite and the host. Unfortunately, the literature contains only a few publications about the use of animals for L. tropica studies. Here, we report an animal model suitable for genetic, pathological and drug studies in L. tropica infection. We show how the host genotype influences different disease symptoms: skin lesions, parasite dissemination to the lymph nodes, spleen and liver, and increase of levels of chemokines CCL2, CCL3 and CCL5 in serum.
Collapse
|
23
|
The Abl and Arg kinases mediate distinct modes of phagocytosis and are required for maximal Leishmania infection. Mol Cell Biol 2012; 32:3176-86. [PMID: 22665498 DOI: 10.1128/mcb.00086-12] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Leishmania, an obligate intracellular parasite, binds several receptors to trigger engulfment by phagocytes, leading to cutaneous or visceral disease. These receptors include complement receptor 3 (CR3), used by promastigotes, and the Fc receptor (FcR), used by amastigotes. The mechanisms mediating uptake are not well understood. Here we show that Abl family kinases mediate both phagocytosis and the uptake of Leishmania amazonensis by macrophages (Ms). Imatinib, an Abl/Arg kinase inhibitor, decreases opsonized polystyrene bead phagocytosis and Leishmania uptake. Interestingly, phagocytosis of IgG-coated beads is decreased in Arg-deficient Ms, while that of C3bi-coated beads is unaffected. Conversely, uptake of C3bi-coated beads is decreased in Abl-deficient Ms, but that of IgG-coated beads is unaffected. Consistent with these results, Abl-deficient Ms are inefficient at C3bi-opsonized promastigote uptake, and Arg-deficient Ms are defective in IgG1-opsonized amastigote uptake. Finally, genetic loss of Abl or Arg reduces infection severity in murine cutaneous leishmaniasis, and imatinib treatment results in smaller lesions with fewer parasites than in controls. Our studies are the first to demonstrate that efficient phagocytosis and maximal Leishmania infection require Abl family kinases. These results highlight Abl family kinase-mediated signaling pathways as potential therapeutic targets for leishmaniasis.
Collapse
|
24
|
Felizardo TC, Gaspar-Elsas MI, Lima GM, Abrahamsohn IA. Lack of signaling by IL-4 or by IL-4/IL-13 has more attenuating effects on Leishmania amazonensis dorsal skin – than on footpad-infected mice. Exp Parasitol 2012; 130:48-57. [DOI: 10.1016/j.exppara.2011.09.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 09/27/2011] [Accepted: 09/30/2011] [Indexed: 10/16/2022]
|
25
|
Castellucci L, Jamieson SE, Miller EN, de Almeida LF, Oliveira J, Magalhães A, Guimarães LH, Lessa M, Lago E, de Jesus AR, Carvalho EM, Blackwell JM. FLI1 polymorphism affects susceptibility to cutaneous leishmaniasis in Brazil. Genes Immun 2011; 12:589-94. [PMID: 21633373 PMCID: PMC3297968 DOI: 10.1038/gene.2011.37] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 02/23/2011] [Indexed: 01/21/2023]
Abstract
Mapping murine genes controlling cutaneous leishmaniasis (CL) identified Fli1 as a candidate influencing resistance to L. major and enhanced wound healing. We examine FLI1 as a gene controlling CL and mucosal leishmaniasis (ML) caused by L. braziliensis in humans. Intron 1 single nucleotide polymorphisms tagging promoter and enhancer elements were analysed in 168 nuclear families (250 CL; 87 ML cases) and replicated in 157 families (402 CL; 39 ML cases). Robust case-pseudocontrol logistic regression analysis showed association between allele C (odds ratio (OR) 1.65; 95% confidence interval 1.18-2.29; P=0.003) of FLI1_rs7930515 and CL in the primary sample that was confirmed (OR 1.60; 95% confidence interval 1.10-2.33; P=0.014) in the replication set (combined P=1.8 × 10(-4)). FLI1_rs7930515 is in linkage disequilibrium with the functional GAn microsatellite in the proximal promoter. Haplotype associations extended across the enhancer, which was not polymorphic. ML associated with inverse haplotypes compared with CL. Wound healing is therefore important in CL, providing potential for therapies modulating FLI1.
Collapse
Affiliation(s)
- L Castellucci
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Federal University of Bahia, Salvador, Brazil
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - SE Jamieson
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Telethon Institute for Child Health Research, Centre for Child Health Research, The University of Western Australia, Subiaco, Western Australia, Australia
| | - EN Miller
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - LF de Almeida
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Federal University of Bahia, Salvador, Brazil
| | - J Oliveira
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Federal University of Bahia, Salvador, Brazil
| | - A Magalhães
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Federal University of Bahia, Salvador, Brazil
| | - LH Guimarães
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Federal University of Bahia, Salvador, Brazil
| | - M Lessa
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Federal University of Bahia, Salvador, Brazil
| | - E Lago
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Federal University of Bahia, Salvador, Brazil
| | - AR de Jesus
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Federal University of Bahia, Salvador, Brazil
- Instituto de Investigação em Imunologia, Faculty of Medicine, University of Sao Paulo, São Paulo, Brazil
- Departmento de Medicina, Universidade Federal de Sergipe, Aracaju, Brazil
| | - EM Carvalho
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Federal University of Bahia, Salvador, Brazil
| | - JM Blackwell
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Telethon Institute for Child Health Research, Centre for Child Health Research, The University of Western Australia, Subiaco, Western Australia, Australia
| |
Collapse
|
26
|
Bujor AM, Asano Y, Haines P, Lafyatis R, Trojanowska M. The c-Abl tyrosine kinase controls protein kinase Cδ-induced Fli-1 phosphorylation in human dermal fibroblasts. ACTA ACUST UNITED AC 2011; 63:1729-37. [PMID: 21321929 DOI: 10.1002/art.30284] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE We have previously demonstrated that in response to transforming growth factor β (TGFβ), Fli-1 activity is repressed through a series of sequential posttranslational modifications, consisting of protein kinase Cδ (PKCδ)-induced Thr312 phosphorylation, acetylation by p300/CREB binding protein-associated factor, and detachment from the collagen promoter. The purpose of this study was to further investigate the upstream events that lead to Fli-1 phosphorylation in response to TGFβ. METHODS Dermal fibroblasts were isolated from systemic sclerosis (SSc) patients and healthy control subjects matched for age, sex, and ethnicity. Western blotting was used to analyze protein levels and real-time quantitative reverse transcription-polymerase chain reaction analysis was used to measure messenger RNA expression. Cells were transduced with constitutively active PKCδ adenovirus or were transiently transfected with a Bcr-Abl-overexpressing plasmid. Subcellular localization of PKCδ was examined by immunocytochemistry. RESULTS Western blot analysis of cell lysates demonstrated that the levels of phospho-Fli-1 (Thr312) were up-regulated in SSc fibroblasts, correlating with increased levels of type I collagen and c-Abl protein. Experiments using a constitutively activated form of c-Abl, small interfering RNA against c-Abl and the specific tyrosine kinase inhibitor imatinib, demonstrated the requirement of c-Abl for the TGFβ-induced phosphorylation of Fli-1. Additionally, we showed that c-Abl kinase activity was required for nuclear localization of PKCδ. CONCLUSION Our results demonstrate that in SSc fibroblasts, c-Abl is an upstream regulator of the profibrotic PKCδ/phospho-Fli-1 pathway, via induction of PKCδ nuclear localization. Additionally, the finding that Fli-1 is phosphorylated at higher levels in SSc fibroblasts supports the notion that the c-Abl/PKCδ/phospho-Fli-1 pathway is constitutively activated in these cells. Thus, blocking the TGFβ/c-Abl/PKCδ/phospho-Fli-1 pathway could be an attractive alternative approach to therapy for scleroderma.
Collapse
Affiliation(s)
- Andreea M Bujor
- Boston University School of Medicine, Arthritis Center-Rheumatology, Boston, Massachusetts 02118, USA
| | | | | | | | | |
Collapse
|
27
|
Abstract
More than 20 years ago, immunologists discovered that resistance and susceptibility to experimental infection with the intracellular protozoan Leishmania major was associated with the development of T-helper 1 (Th1)- and Th2-dominated immune responses, respectively. This infectious disease model was later used to identify and assess the role of key factors, such as interleukin-12 (IL-12) and IL-4, in Th1 and Th2 maturation. While infection by Leishmania remains a popular model for immunologists who wish to assess the role of their favorite molecule in T-cell differentiation, other investigators have tried to better understand how Leishmania interact with its insect and mammalian hosts. In this review, we discuss some of these new data with an emphasis on the early events that shape the immune response to Leishmania and on the immune evasion mechanisms that allow this parasite to avoid the development of sterilizing immunity and to secure its transmission to a new host.
Collapse
Affiliation(s)
- Evelyne Mougneau
- Institut National de la Santé et de la Recherche Médicale, University of Nice-Sophia Antipolis, Valbonne, France
| | | | | |
Collapse
|
28
|
Hattori T, Stawski L, Nakerakanti SS, Trojanowska M. Fli1 is a negative regulator of estrogen receptor α in dermal fibroblasts. J Invest Dermatol 2011; 131:1469-76. [PMID: 21451544 DOI: 10.1038/jid.2011.63] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Estrogen is an important regulator of dermal fibroblast functions, including extracellular matrix (ECM) synthesis. Estrogen mediates its effects through estrogen receptors (ERs), ERα and ERβ; however, regulation of ERs in dermal fibroblasts remains poorly understood. Friend leukemia integration factor 1 (Fli1), a member of the Ets transcription factor family, has been shown to play a pivotal role in regulation of the ECM genes in dermal fibroblasts. The aim of this study was to examine a possible interaction between Fli1 and estrogen pathways, focusing on ERα. We show that treatment of human dermal fibroblasts with transforming growth factor-β (TGF-β) increases ERα protein and mRNA levels. Similarly, ERα expression was increased in response to small interfering RNA (siRNA)-mediated depletion of Fli1, suggesting that Fli1 is a mediator of the TGF-β effects on ERα expression. Accordingly, we showed that Fli1 binds to the most proximal region of the ERα promoter, and dissociates from the promoter upon TGF-β treatment. An inverse correlation between Fli1 and ERα expression levels was confirmed in cultured skin fibroblasts obtained from Fli1(+/-) mice and in the skin of Fli1(+/-) mice in vivo. This study supports a role of Fli1 as a negative regulator of the ERα gene in dermal fibroblasts.
Collapse
Affiliation(s)
- Tomoyasu Hattori
- Arthritis Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
29
|
Van Der Kraak L, Meunier C, Turbide C, Jothy S, Gaboury L, Marcus V, Chang SY, Beauchemin N, Gros P. A two-locus system controls susceptibility to colitis-associated colon cancer in mice. Oncotarget 2010; 1:436-446. [PMID: 21311099 PMCID: PMC3248117 DOI: 10.18632/oncotarget.177] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Accepted: 10/10/2010] [Indexed: 11/25/2022] Open
Abstract
We have previously shown that the differential susceptibility of A/J (susceptible) and C57BL/6J (B6, resistant) mouse strains to azoxymethane (AOM)-induced colorectal cancer (CRC) is controlled by the chromosome 3 locus, Ccs3. We report that A/J and B6 mice also show differential susceptibility to colitis-associated colorectal cancer (CA-CRC) induced by combined administration of AOM and dextran sulfate. This differential susceptibility is not controlled by Ccs3, but is under distinct genetic control. Linkage analyses in (A/J x B6)F2 mice detected a major CA-CRC susceptibility locus on chromosome 9 (Ccs4) which controls tumor multiplicity and tumor surface area. Susceptibility alleles at Ccs4 are inherited in a recessive fashion, with A/J alleles being associated with susceptibility. We also detected a second locus on chromosome 14 that acts in an additive fashion with Ccs4. Strikingly, F2 mice homozygous for A/J alleles at both loci (Ccs4 and chromosome 14) are as susceptible to CA-CRC as the A/J controls while mice homozygous for B6 alleles are as resistant as the B6 controls, thus supporting the role of two interacting loci in this CA-CRC model. This indicates that susceptibility to chemically-induced CRC and susceptibility to CA-CRC are under distinct genetic control in mice, and probably involve distinct cellular pathways.
Collapse
Affiliation(s)
- Lauren Van Der Kraak
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Charles Meunier
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Claire Turbide
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Serge Jothy
- Department of Laboratory Medicine and Pathobiology, St. Michael's Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Louis Gaboury
- Institut de Recherche en Immunologie et en Cancérologie, Université de Montréal, Montreal, Quebec, Canada
| | - Victoria Marcus
- Department of Pathology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Sing Yun Chang
- Department of Pathology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Nicole Beauchemin
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
- Departments of Medicine and Oncology, McGill University, Montreal, Quebec, Canada
| | - Philippe Gros
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| |
Collapse
|