1
|
Bhattacharya S, Chakraborty S, Manna D, Thakur P, Chakravorty N, Mukherjee B. Deciphering the intricate dynamics of inflammasome regulation in visceral and post-kala-azar dermal leishmaniasis: A meta-analysis of consistencies. Acta Trop 2024; 257:107313. [PMID: 38964632 DOI: 10.1016/j.actatropica.2024.107313] [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: 04/11/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Post Kala-azar dermal leishmaniasis (PKDL) arises as a significant dermal sequel following Visceral leishmaniasis (VL) caused by protozoan parasite Leishmania donovani (LD). PKDL acts as a significant constrain for VL elimination serving as a crucial reservoir for LD. PKDL patients exhibit depigmented macular and papular lesions on their skin, which results in social discrimination due to loss of natural skin color. Inflammatory reactions, prevalent in both VL and PKDL, potentially lead to tissue damage in areas harboring the parasite. Disruption of the immune-inflammasomal network not only facilitates LD persistence but also leads to the skin hypopigmentation seen in PKDL, impacting social well-being. Activation of inflammasomal markers like STAT1, NLRP1, NLRP3, AIM2, CASP11, and NLRP12 have been identified as a common host-defense mechanism across various Leishmania infections. Conversely, Leishmania modulates inflammasome activation to sustain its presence within the host. Nevertheless, in specific instances of Leishmania infection, inflammasome activation can worsen disease pathology by promoting parasite proliferation and persistence. This study encompasses recent transcriptomic analyses conducted between 2016 and 2023 on human and murine subjects afflicted with VL/PKDL, elucidating significant alterations in inflammasomal markers in both conditions. It offers a comprehensive understanding how these markers contribute in disease progression, drawing upon available literature for logical analysis. Furthermore, our analysis identifies validated miRNA network that could potentially disrupt this crucial immune-inflammasomal network, thereby offering a plausible explanation on how secreted LD-factors could enable membrane-bound LD, isolated from the host cytoplasm, to modulate cytoplasmic inflammasomal markers. Insights from this study could guide the development of host-directed therapeutics to impede transmission and address hypopigmentation, thereby mitigating the social stigma associated with PKDL.
Collapse
Affiliation(s)
| | | | - Debolina Manna
- School of Medical Science and Technology, IIT Kharagpur, Kharagpur 721302, India
| | - Pradipti Thakur
- School of Medical Science and Technology, IIT Kharagpur, Kharagpur 721302, India
| | - Nishant Chakravorty
- School of Medical Science and Technology, IIT Kharagpur, Kharagpur 721302, India
| | - Budhaditya Mukherjee
- School of Medical Science and Technology, IIT Kharagpur, Kharagpur 721302, India.
| |
Collapse
|
2
|
Takamiya NT, Rogerio LA, Torres C, Leonel JAF, Vioti G, de Sousa Oliveira TMF, Valeriano KC, Porcino GN, de Miranda Santos IKF, Costa CHN, Costa DL, Ferreira TS, Gurgel-Gonçalves R, da Silva JS, Teixeira FR, De Almeida RP, Ribeiro JMC, Maruyama SR. Parasite Detection in Visceral Leishmaniasis Samples by Dye-Based qPCR Using New Gene Targets of Leishmania infantum and Crithidia. Trop Med Infect Dis 2023; 8:405. [PMID: 37624343 PMCID: PMC10457869 DOI: 10.3390/tropicalmed8080405] [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: 07/03/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023] Open
Abstract
Visceral leishmaniasis (VL) is a neglected disease considered a serious public health problem, especially in endemic countries. Several studies have discovered monoxenous trypanosomatids (Leptomonas and Crithidia) in patients with VL. In different situations of leishmaniasis, investigations have examined cases of co-infection between Leishmania spp. and Crithidia spp. These coinfections have been observed in a wide range of vertebrate hosts, indicating that they are not rare. Diagnostic techniques require improvements and more robust tools to accurately detect the causative agent of VL. This study aimed to develop a real-time quantitative dye-based PCR (qPCR) assay capable of distinguishing Leishmania infantum from Crithidia-related species and to estimate the parasite load in samples of VL from humans and animals. The primer LinJ31_2420 targets an exclusive phosphatase of L. infantum; the primer Catalase_LVH60-12060_1F targets the catalase gene of Crithidia. Therefore, primers were designed to detect L. infantum and Crithidia sp. LVH60A (a novel trypanosomatid isolated from VL patients in Brazil), in samples related to VL. These primers were considered species-specific, based on sequence analysis using genome data retrieved from the TriTryp database and the genome assembling of Crithidia sp. LVH60A strain, in addition to experimental and clinical data presented herein. This novel qPCR assay was highly accurate in identifying and quantifying L. infantum and Crithidia sp. LVH60A in samples obtained experimentally (in vitro and in vivo) or collected from hosts (humans, dogs, cats, and vectors). Importantly, the screening of 62 cultured isolates from VL patients using these primers surprisingly revealed that 51 parasite cultures were PCR+ for Crithidia sp. In addition, qPCR assays identified the co-infection of L. infantum with Crithidia sp. LVH60A in two new VL cases in Brazil, confirming the suspicion of co-infection in a previously reported case of fatal VL. We believe that the species-specific genes targeted in this study can be helpful for the molecular diagnosis of VL, as well as for elucidating suspected co-infections with monoxenous-like trypanosomatids, which is a neglected fact of a neglected disease.
Collapse
Affiliation(s)
- Nayore Tamie Takamiya
- Department of Genetics and Evolution, Center for Biological Sciences and Health, Federal University of São Carlos (UFSCar), São Carlos 13565-905, SP, Brazil; (N.T.T.); (F.R.T.)
| | - Luana Aparecida Rogerio
- Department of Genetics and Evolution, Center for Biological Sciences and Health, Federal University of São Carlos (UFSCar), São Carlos 13565-905, SP, Brazil; (N.T.T.); (F.R.T.)
| | - Caroline Torres
- Department of Genetics and Evolution, Center for Biological Sciences and Health, Federal University of São Carlos (UFSCar), São Carlos 13565-905, SP, Brazil; (N.T.T.); (F.R.T.)
| | - João Augusto Franco Leonel
- Post-Graduate Program in Experimental Epidemiology Applied to Zoonoses at the Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-270, SP, Brazil
| | - Geovanna Vioti
- Post-Graduate Program in Experimental Epidemiology Applied to Zoonoses at the Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-270, SP, Brazil
| | - Tricia Maria Ferreira de Sousa Oliveira
- Post-Graduate Program in Experimental Epidemiology Applied to Zoonoses at the Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-270, SP, Brazil
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga 13635-900, SP, Brazil
| | - Karoline Camila Valeriano
- Ribeirão Preto Medical School, University of São Paulo, FMRP-USP, Ribeirão Preto 14049-900, SP, Brazil
| | | | | | - Carlos H. N. Costa
- Natan Portela Institute of Tropical Diseases, Teresina 64002-510, PI, Brazil
| | | | - Tauana Sousa Ferreira
- Laboratory of Medical Parasitology and Vector Biology, Faculty of Medicine, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Rodrigo Gurgel-Gonçalves
- Laboratory of Medical Parasitology and Vector Biology, Faculty of Medicine, University of Brasília, Brasília 70910-900, DF, Brazil
| | - João Santana da Silva
- Fiocruz-Bi-Institutional Translational Medicine Project, Oswaldo Cruz Foundation, Ribeirão Preto 14040-900, SP, Brazil
| | - Felipe Roberti Teixeira
- Department of Genetics and Evolution, Center for Biological Sciences and Health, Federal University of São Carlos (UFSCar), São Carlos 13565-905, SP, Brazil; (N.T.T.); (F.R.T.)
| | - Roque Pacheco De Almeida
- Department of Medicine, Center for Biology and Health Sciences, Federal University of Sergipe (UFS), Aracaju 49060-108, SE, Brazil
| | - José M. C. Ribeiro
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, NIH/NIAID, Rockville, MD 20892, USA
| | - Sandra Regina Maruyama
- Department of Genetics and Evolution, Center for Biological Sciences and Health, Federal University of São Carlos (UFSCar), São Carlos 13565-905, SP, Brazil; (N.T.T.); (F.R.T.)
| |
Collapse
|
3
|
Porcino GN, Bladergroen MR, Dotz V, Nicolardi S, Memarian E, Gardinassi LG, Nery Costa CH, Pacheco de Almeida R, Ferreira de Miranda Santos IK, Wuhrer M. Total serum N-glycans mark visceral leishmaniasis in human infections with Leishmania infantum. iScience 2023; 26:107021. [PMID: 37485378 PMCID: PMC10362369 DOI: 10.1016/j.isci.2023.107021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/07/2023] [Accepted: 05/30/2023] [Indexed: 07/25/2023] Open
Abstract
Visceral leishmaniasis (VL) is a clinical form of leishmaniasis with high mortality rates when not treated. Diagnosis suffers from invasive techniques and sub-optimal sensitivities. The current (affordable) treatment with pentavalent antimony as advised by the WHO is possibly harmful to the patient. There is need for an improved diagnosis to prevent possibly unnecessary treatment. N-glycan analysis may aid in diagnosis. We evaluated the N-glycan profiles from active VL, asymptomatic infections (ASYMP) and controls from non-endemic (NC) and endemic (EC) areas. Active VL has a distinct N-glycome profile that associates with disease severity. Our study suggests that the observed glycan signatures could be a valuable additive to diagnosis and assist in identifying possible markers of disease and understanding the pathogenesis of VL. Further studies are warranted to assess a possible future role of blood glycome analysis in active VL diagnosis and should aim at disease specificity.
Collapse
Affiliation(s)
- Gabriane Nascimento Porcino
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Marco René Bladergroen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Viktoria Dotz
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Simone Nicolardi
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Elham Memarian
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Luiz Gustavo Gardinassi
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, Brazil
| | | | - Roque Pacheco de Almeida
- Departamento de Medicina, Programa de Pós-Graduação em Ciências da Saúde – PPGCS, Universidade Federal de Sergipe, Aracajú 49060-100, Brazil
| | | | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| |
Collapse
|
4
|
de Moraes L, Santos LA, Arruda LB, da Silva MDPP, Silva MDO, Silva JAG, Ramos A, dos Santos MB, Torres FG, Orge C, Teixeira AMDS, Vieira TS, Ramírez L, Soto M, Grassi MFR, de Siqueira IC, Costa DL, Costa CHN, Andrade BDB, Akrami K, de Oliveira CI, Boaventura VS, Barral-Netto M, Barral A, Vandamme AM, Van Weyenbergh J, Khouri R. High seroprevalence of Leishmania infantum is linked to immune activation in people with HIV: a two-stage cross-sectional study in Bahia, Brazil. Front Microbiol 2023; 14:1221682. [PMID: 37601355 PMCID: PMC10436095 DOI: 10.3389/fmicb.2023.1221682] [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: 05/12/2023] [Accepted: 06/28/2023] [Indexed: 08/22/2023] Open
Abstract
Visceral leishmaniasis is an opportunistic disease in HIV-1 infected individuals, unrecognized as a determining factor for AIDS diagnosis. The growing geographical overlap of HIV-1 and Leishmania infections is an emerging challenge worldwide, as co-infection increases morbidity and mortality for both infections. Here, we determined the prevalence of people living with HIV (PWH) with a previous or ongoing infection by Leishmania infantum and investigated the virological and immunological factors associated with co-infection. We adopted a two-stage cross-sectional cohort (CSC) design (CSC-I, n = 5,346 and CSC-II, n = 317) of treatment-naïve HIV-1-infected individuals in Bahia, Brazil. In CSC-I, samples collected between 1998 and 2013 were used for serological screening for leishmaniasis by an in-house Enzyme-Linked Immunosorbent Assay (ELISA) with SLA (Soluble Leishmania infantum Antigen), resulting in a prevalence of previous or ongoing infection of 16.27%. Next, 317 PWH were prospectively recruited from July 2014 to December 2015 with the collection of sociodemographic and clinical data. Serological validation by two different immunoassays confirmed a prevalence of 15.46 and 8.20% by anti-SLA, and anti-HSP70 serology, respectively, whereas 4.73% were double-positive (DP). Stratification of these 317 individuals in DP and double-negative (DN) revealed a significant reduction of CD4+ counts and CD4+/CD8+ ratios and a tendency of increased viral load in the DP group, as compared to DN. No statistical differences in HIV-1 subtype distribution were observed between the two groups. However, we found a significant increase of CXCL10 (p = 0.0076) and a tendency of increased CXCL9 (p = 0.061) in individuals with DP serology, demonstrating intensified immune activation in this group. These findings were corroborated at the transcriptome level in independent Leishmania- and HIV-1-infected cohorts (Swiss HIV Cohort and Piaui Northeast Brazil Cohort), indicating that CXCL10 transcripts are shared by the IFN-dominated immune activation gene signatures of both pathogens and positively correlated to viral load in untreated PWH. This study demonstrated a high prevalence of PWH with L. infantum seropositivity in Bahia, Brazil, linked to IFN-mediated immune activation and a significant decrease in CD4+ levels. Our results highlight the urgent need to increase awareness and define public health strategies for the management and prevention of HIV-1 and L. infantum co-infection.
Collapse
Affiliation(s)
- Laise de Moraes
- Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Luciane Amorim Santos
- Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
| | - Liã Bárbara Arruda
- Centre for Clinical Microbiology, Division of Infection & Immunity, University College London, London, United Kingdom
| | | | - Márcio de Oliveira Silva
- Centro Estadual Especializado em Diagnóstico, Assistência e Pesquisa, Secretaria de Saúde do Estado da Bahia, Salvador, Brazil
| | - José Adriano Góes Silva
- Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Centro Estadual Especializado em Diagnóstico, Assistência e Pesquisa, Secretaria de Saúde do Estado da Bahia, Salvador, Brazil
| | - André Ramos
- Centro Estadual Especializado em Diagnóstico, Assistência e Pesquisa, Secretaria de Saúde do Estado da Bahia, Salvador, Brazil
| | | | | | - Cibele Orge
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | | | | | - Laura Ramírez
- Departamento de Biología Molecular, Facultad de Ciencias, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Soto
- Departamento de Biología Molecular, Facultad de Ciencias, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Maria Fernanda Rios Grassi
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
| | | | - Dorcas Lamounier Costa
- Laboratório de Leishmanioses, Instituto de Doenças Tropicais Natan Portella, Universidade Federal do Piauí, Teresina, Brazil
| | - Carlos Henrique Nery Costa
- Laboratório de Leishmanioses, Instituto de Doenças Tropicais Natan Portella, Universidade Federal do Piauí, Teresina, Brazil
| | - Bruno de Bezerril Andrade
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
| | - Kevan Akrami
- Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Camila Indiani de Oliveira
- Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
| | - Viviane Sampaio Boaventura
- Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Hospital Santa Izabel, Salvador, Brazil
| | - Manoel Barral-Netto
- Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Aldina Barral
- Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Anne-Mieke Vandamme
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
- Center for Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Johan Van Weyenbergh
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
| | - Ricardo Khouri
- Programa de Pós-graduação em Ciências da Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador, Brazil
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
| |
Collapse
|
5
|
da Matta VLR, Gonçalves AN, Gomes CMC, Chouman IH, Ferreira FM, Campos MB, Lima LV, Vasconcelos dos Santos T, Ramos PK, Furtado RR, Laurenti MD, Corbett CEP, Nakaya HI, Silveira FT. Gene Signatures of Symptomatic and Asymptomatic Clinical-Immunological Profiles of Human Infection by Leishmania (L.) chagasi in Amazonian Brazil. Microorganisms 2023; 11:microorganisms11030653. [PMID: 36985226 PMCID: PMC10058599 DOI: 10.3390/microorganisms11030653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/18/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Individuals infected with Leishmania (L.) chagasi may present different asymptomatic and symptomatic stages of infection, which vary in the clinical–immunological profiles that can be classified as asymptomatic infection (AI), subclinical resistant infection (SRI), indeterminate initial infection (III), subclinical oligosymptomatic infection (SOI), and symptomatic infection (SI) (=American visceral leishmaniasis, AVL). However, little is known about the molecular differences between individuals having each profile. Here, we performed whole-blood transcriptomic analyses of 56 infected individuals from Pará State (Brazilian Amazon), covering all five profiles. We then identified the gene signatures of each profile by comparing their transcriptome with those of 11 healthy individuals from the same area. Symptomatic individuals with SI (=AVL) and SOI profiles showed higher transcriptome perturbation when compared to those asymptomatic III, AI and SRI profiles, suggesting that disease severity may be associated with greater transcriptomic changes. Although the expression of many genes was altered on each profile, very few genes were shared among the profiles. This indicated that each profile has a unique gene signature. The innate immune system pathway was strongly activated only in asymptomatic AI and SRI profiles, suggesting the control of infection. In turn, pathways such as MHC Class II antigen presentation and NF-kB activation in B cells seemed to be specifically induced in symptomatic SI (=AVL) and SOI profiles. Moreover, cellular response to starvation was down-regulated in those symptomatic profiles. Overall, this study revealed five distinct transcriptional patterns associated to the clinical–immunological (symptomatic and asymptomatic) profiles of human L. (L.) chagasi-infection in the Brazilian Amazon.
Collapse
Affiliation(s)
- Vania Lucia R. da Matta
- Laboratorio de Patologia de Molestias Infecciosas (LIM-50), Departamento de Patologia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01246 903, Brazil
| | - André N. Gonçalves
- Laboratorio de Patologia de Molestias Infecciosas (LIM-50), Departamento de Patologia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01246 903, Brazil
| | - Cláudia Maria C. Gomes
- Laboratorio de Patologia de Molestias Infecciosas (LIM-50), Departamento de Patologia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01246 903, Brazil
| | - Islam H. Chouman
- Laboratorio de Patologia de Molestias Infecciosas (LIM-50), Departamento de Patologia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01246 903, Brazil
| | - Frederico M. Ferreira
- Laboratorio de Patologia de Molestias Infecciosas (LIM-50), Departamento de Patologia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01246 903, Brazil
| | - Marliane B. Campos
- Parasitology Department, Evandro Chagas Institute, Health Surveillance Secretary, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Luciana V. Lima
- Parasitology Department, Evandro Chagas Institute, Health Surveillance Secretary, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Thiago Vasconcelos dos Santos
- Parasitology Department, Evandro Chagas Institute, Health Surveillance Secretary, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Patrícia Karla Ramos
- Parasitology Department, Evandro Chagas Institute, Health Surveillance Secretary, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Rodrigo R. Furtado
- Parasitology Department, Evandro Chagas Institute, Health Surveillance Secretary, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Marcia D. Laurenti
- Laboratorio de Patologia de Molestias Infecciosas (LIM-50), Departamento de Patologia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01246 903, Brazil
| | - Carlos Eduardo P. Corbett
- Laboratorio de Patologia de Molestias Infecciosas (LIM-50), Departamento de Patologia, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01246 903, Brazil
| | - Helder I. Nakaya
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciencias Farmaceuticas, Universidade de Sao Paulo, Sao Paulo 05508-220, Brazil
- Hospital Israelita Albert Einstein, Sao Paulo 05652-900, Brazil
- Correspondence: (H.I.N.); (F.T.S.)
| | - Fernando T. Silveira
- Parasitology Department, Evandro Chagas Institute, Health Surveillance Secretary, Ministry of Health, Ananindeua 67030-000, Brazil
- Tropical Medicine Nucleus, Pará Federal University, Belém 67030-000, Brazil
- Correspondence: (H.I.N.); (F.T.S.)
| |
Collapse
|
6
|
Saidi N, Jelassi R. Does infection with Leishmania protect against Covid-19? Immunol Lett 2023; 253:28-29. [PMID: 36603800 PMCID: PMC9807266 DOI: 10.1016/j.imlet.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/29/2022] [Accepted: 01/02/2023] [Indexed: 01/04/2023]
Affiliation(s)
- Nasreddine Saidi
- Laboratoire de Recherche, Parasitoses Médicales, Biotechnologies et Biomolécules, LR 20-IPT-06, Institut Pasteur de Tunis, Université Tunis El-Manar, Tunis 1002, Tunisia.
| | | |
Collapse
|
7
|
Bekkar A, Isorce N, Snäkä T, Claudinot S, Desponds C, Kopelyanskiy D, Prével F, Reverte M, Xenarios I, Fasel N, Teixeira F. Dissection of the macrophage response towards infection by the Leishmania-viral endosymbiont duo and dynamics of the type I interferon response. Front Cell Infect Microbiol 2022; 12:941888. [PMID: 35992159 PMCID: PMC9386148 DOI: 10.3389/fcimb.2022.941888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/01/2022] [Indexed: 11/25/2022] Open
Abstract
Leishmania RNA virus 1 (LRV1) is a double-stranded RNA virus found in some strains of the human protozoan parasite Leishmania, the causative agent of leishmaniasis, a neglected tropical disease. Interestingly, the presence of LRV1 inside Leishmania constitutes an important virulence factor that worsens the leishmaniasis outcome in a type I interferon (IFN)–dependent manner and contributes to treatment failure. Understanding how macrophages respond toward Leishmania alone or in combination with LRV1 as well as the role that type I IFNs may play during infection is fundamental to oversee new therapeutic strategies. To dissect the macrophage response toward infection, RNA sequencing was performed on murine wild-type and Ifnar-deficient bone marrow–derived macrophages infected with Leishmania guyanensis (Lgy) devoid or not of LRV1. Additionally, macrophages were treated with poly I:C (mimetic virus) or with type I IFNs. By implementing a weighted gene correlation network analysis, the groups of genes (modules) with similar expression patterns, for example, functionally related, coregulated, or the members of the same functional pathway, were identified. These modules followed patterns dependent on Leishmania, LRV1, or Leishmania exacerbated by the presence of LRV1. Not only the visualization of how individual genes were embedded to form modules but also how different modules were related to each other were observed. Thus, in the context of the observed hyperinflammatory phenotype associated to the presence of LRV1, it was noted that the biomarkers tumor-necrosis factor α (TNF-α) and the interleukin 6 (IL-6) belonged to different modules and that their regulating specific Src-family kinases were segregated oppositely. In addition, this network approach revealed the strong and sustained effect of LRV1 on the macrophage response and genes that had an early, late, or sustained impact during infection, uncovering the dynamics of the IFN response. Overall, this study contributed to shed light and dissect the intricate macrophage response toward infection by the Leishmania-LRV1 duo and revealed the crosstalk between modules made of coregulated genes and provided a new resource that can be further explored to study the impact of Leishmania on the macrophage response.
Collapse
Affiliation(s)
- Amel Bekkar
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Nathalie Isorce
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Tiia Snäkä
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | | | - Chantal Desponds
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | | | - Florence Prével
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Marta Reverte
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Ioannis Xenarios
- Agora Center, Center Hospitalier Universitaire (CHUV), Lausanne, Switzerland
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Nicolas Fasel
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- *Correspondence: Nicolas Fasel, ; Filipa Teixeira,
| | - Filipa Teixeira
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- *Correspondence: Nicolas Fasel, ; Filipa Teixeira,
| |
Collapse
|
8
|
Bahrami F, Masoudzadeh N, Van Veen S, Persson J, Lari A, Sarvnaz H, Taslimi Y, Östensson M, Andersson B, Sharifi I, Goyonlo VM, Ottenhoff TH, Haks MC, Harandi AM, Rafati S. Blood transcriptional profiles distinguish different clinical stages of cutaneous leishmaniasis in humans. Mol Immunol 2022; 149:165-173. [PMID: 35905592 DOI: 10.1016/j.molimm.2022.07.008] [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: 03/27/2022] [Revised: 07/03/2022] [Accepted: 07/19/2022] [Indexed: 10/16/2022]
Abstract
Cutaneous leishmaniasis (CL) is a neglected tropical disease with severe morbidity and socioeconomic sequelae. A better understanding of underlying immune mechanisms that lead to different clinical outcomes of CL could inform the rational design of intervention measures. While transcriptomic analyses of CL lesions were recently reported by us and others, there is a dearth of information on the expression of immune-related genes in the blood of CL patients. Herein, we investigated immune-related gene expression in whole blood samples collected from individuals with different clinical stages of CL along with healthy volunteers in an endemic CL region where Leishmania (L.) tropica is prevalent. Study participants were categorized into asymptomatic (LST+) and healthy uninfected (LST-) groups based on their leishmanin skin test (LST). Whole blood PAXgene samples were collected from volunteers, who had healed CL lesions, and patients with active L. tropica cutaneous lesions. Quality RNA extracted from 57 blood samples were subjected to Dual-color reverse-transcription multiplex ligation-dependent probe amplification (dcRT-MLPA) assay for profiling 144 immune-related genes. Results show significant changes in the expression of genes involved in interferon signaling pathway in the blood of active CL patients, asymptomatics and healed individuals. Nonetheless, distinct profiles for several immune-related genes were identified in the healed, the asymptomatic, and the CL patients compared to the healthy controls. Among others, IFI16 and CCL11 were found as immune transcript signatures for the healed and the asymptomatic individuals, respectively. These results warrant further exploration to pinpoint novel blood biomarkers for different clinical stages of CL.
Collapse
Affiliation(s)
- Fariborz Bahrami
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Nasrin Masoudzadeh
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Suzanne Van Veen
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Josefine Persson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Arezou Lari
- Systems Biomedicine Unit, Pasteur Institute of Iran, Tehran, Iran
| | - Hamzeh Sarvnaz
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Yasaman Taslimi
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| | - Malin Östensson
- Bioinformatics Core Facility, University of Gothenburg, Gothenburg, Sweden
| | - Björn Andersson
- Bioinformatics Core Facility, University of Gothenburg, Gothenburg, Sweden
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Tom Hm Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Mariëlle C Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Ali M Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Vaccine Evaluation Center, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada.
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
9
|
Dirkx L, Hendrickx S, Merlot M, Bulté D, Starick M, Elst J, Bafica A, Ebo DG, Maes L, Van Weyenbergh J, Caljon G. Long-term hematopoietic stem cells as a parasite niche during treatment failure in visceral leishmaniasis. Commun Biol 2022; 5:626. [PMID: 35752645 PMCID: PMC9233693 DOI: 10.1038/s42003-022-03591-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/14/2022] [Indexed: 01/20/2023] Open
Abstract
Given the discontinuation of various first-line drugs for visceral leishmaniasis (VL), large-scale in vivo drug screening, establishment of a relapse model in rodents, immunophenotyping, and transcriptomics were combined to study persistent infections and therapeutic failure. Double bioluminescent/fluorescent Leishmania infantum and L. donovani reporter lines enabled the identification of long-term hematopoietic stem cells (LT-HSC) as a niche in the bone marrow with remarkably high parasite burdens, a feature confirmed for human hematopoietic stem cells (hHSPC). LT-HSC are more tolerant to antileishmanial drug action and serve as source of relapse. A unique transcriptional ’StemLeish’ signature in these cells was defined by upregulated TNF/NF-κB and RGS1/TGF-β/SMAD/SKIL signaling, and a downregulated oxidative burst. Cross-species analyses demonstrated significant overlap with human VL and HIV co-infected blood transcriptomes. In summary, the identification of LT-HSC as a drug- and oxidative stress-resistant niche, undergoing a conserved transcriptional reprogramming underlying Leishmania persistence and treatment failure, may open therapeutic avenues for leishmaniasis. Long-term hematopoietic stem cells may act as protective niches for the Leishmania parasite, potentially contributing to treatment failure in cases of visceral leishmaniasis.
Collapse
Affiliation(s)
- Laura Dirkx
- Laboratory of Microbiology, Parasitology, and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Sarah Hendrickx
- Laboratory of Microbiology, Parasitology, and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Margot Merlot
- Laboratory of Microbiology, Parasitology, and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Dimitri Bulté
- Laboratory of Microbiology, Parasitology, and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Marick Starick
- Clinical and Epidemiological Virology, Department of Microbiology, Immunology, and Transplantation, Rega Institute of Medical Research, KU Leuven, Leuven, Belgium.,Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology Federal University of Santa Catarina, Florianopolis, Brazil
| | - Jessy Elst
- Department of Immunology-Allergology-Rheumatology, Faculty of Medicine and Health Science and the Infla-Med Centre of Excellence, University of Antwerp, Antwerp University Hospital, Antwerp, Belgium
| | - André Bafica
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology Federal University of Santa Catarina, Florianopolis, Brazil
| | - Didier G Ebo
- Department of Immunology-Allergology-Rheumatology, Faculty of Medicine and Health Science and the Infla-Med Centre of Excellence, University of Antwerp, Antwerp University Hospital, Antwerp, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology, and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Johan Van Weyenbergh
- Clinical and Epidemiological Virology, Department of Microbiology, Immunology, and Transplantation, Rega Institute of Medical Research, KU Leuven, Leuven, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology, and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
10
|
Snäkä T, Bekkar A, Desponds C, Prével F, Claudinot S, Isorce N, Teixeira F, Grasset C, Xenarios I, Lopez-Mejia IC, Fajas L, Fasel N. Sex-Biased Control of Inflammation and Metabolism by a Mitochondrial Nod-Like Receptor. Front Immunol 2022; 13:882867. [PMID: 35651602 PMCID: PMC9150262 DOI: 10.3389/fimmu.2022.882867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/05/2022] [Indexed: 12/17/2022] Open
Abstract
Mitochondria regulate steroid hormone synthesis, and in turn sex hormones regulate mitochondrial function for maintaining cellular homeostasis and controlling inflammation. This crosstalk can explain sex differences observed in several pathologies such as in metabolic or inflammatory disorders. Nod-like receptor X1 (NLRX1) is a mitochondria-associated innate receptor that could modulate metabolic functions and attenuates inflammatory responses. Here, we showed that in an infectious model with the human protozoan parasite, Leishmania guyanensis, NLRX1 attenuated inflammation in females but not in male mice. Analysis of infected female and male bone marrow derived macrophages showed both sex- and genotype-specific differences in both inflammatory and metabolic profiles with increased type I interferon production, mitochondrial respiration, and glycolytic rate in Nlrx1-deficient female BMDMs in comparison to wild-type cells, while no differences were observed between males. Transcriptomics of female and male BMDMs revealed an altered steroid hormone signaling in Nlrx1-deficient cells, and a “masculinization” of Nlrx1-deficient female BMDMs. Thus, our findings suggest that NLRX1 prevents uncontrolled inflammation and metabolism in females and therefore may contribute to the sex differences observed in infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Tiia Snäkä
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Amel Bekkar
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Chantal Desponds
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Florence Prével
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | | | - Nathalie Isorce
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Filipa Teixeira
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Coline Grasset
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Ioannis Xenarios
- Agora Center, Center Hospitalier Universitaire (CHUV), Lausanne, Switzerland.,Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | | | - Lluis Fajas
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Nicolas Fasel
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
11
|
Forrester S, Goundry A, Dias BT, Leal-Calvo T, Moraes MO, Kaye PM, Mottram JC, Lima APCA. Tissue Specific Dual RNA-Seq Defines Host-Parasite Interplay in Murine Visceral Leishmaniasis Caused by Leishmania donovani and Leishmania infantum. Microbiol Spectr 2022; 10:e0067922. [PMID: 35384718 PMCID: PMC9045295 DOI: 10.1128/spectrum.00679-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 01/22/2023] Open
Abstract
Visceral leishmaniasis is associated with hepato-splenomegaly and altered immune and hematological parameters in both preclinical animal models and humans. We studied mouse experimental visceral leishmaniasis caused by Leishmania infantum and Leishmania donovani in BALB/c mice using dual RNA-seq to investigate the transcriptional response of host and parasite in liver and spleen. We identified only 4 species-specific parasite expressed genes (SSPEGs; log2FC >1, FDR <0.05) in the infected spleen, and none in the infected liver. For the host transcriptome, we found 789 differentially expressed genes (DEGs; log2FC >1, FDR <0.05) in the spleen that were common to both infections, with IFNγ signaling and complement and coagulation cascade pathways highly enriched, and an additional 286 and 186 DEGs that were selective to L. donovani and L. infantum infection, respectively. Among those, there were network interactions between genes of amino acid metabolism and PPAR signaling in L. donovani infection and increased IL1β and positive regulation of fatty acid transport in L. infantum infection, although no pathway enrichment was observed. In the liver, there were 1,939 DEGs in mice infected with either L. infantum or L. donovani in comparison to uninfected mice, and the most enriched pathways were IFNγ signaling, neutrophil mediated immunity, complement and coagulation, cytokine-chemokine responses, and hemostasis. Additionally, 221 DEGs were selective in L. donovani and 429 DEGs in L. infantum infections. These data show that the host response for these two visceral leishmaniasis infection models is broadly similar, and ∼10% of host DEGs vary in infections with either parasite species. IMPORTANCE Visceral leishmaniasis (VL) is caused by two species of Leishmania parasites, L. donovani in the Old World and L. infantum in the New World and countries bordering the Mediterranean. Although cardinal features such as hepato-splenomegaly and alterations in blood and immune function are evident, clinical presentation may vary by geography, with for example severe bleeding often associated with VL in Brazil. Although animal models of both L. donovani and L. infantum have been widely used to study disease pathogenesis, a direct side-by-side comparison of how these parasites species impact the infected host and/or how they might respond to the stresses of mammalian infection has not been previously reported. Identifying common and distinct pathways to pathogenesis will be important to ensure that new therapeutic or prophylactic approaches will be applicable across all forms of VL.
Collapse
Affiliation(s)
- Sarah Forrester
- York Biomedical Research Institute, Department of Biology, University of York, York, England, United Kingdom
| | - Amy Goundry
- York Biomedical Research Institute, Department of Biology, University of York, York, England, United Kingdom
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruna Torres Dias
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Paul M. Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, England, United Kingdom
| | - Jeremy C. Mottram
- York Biomedical Research Institute, Department of Biology, University of York, York, England, United Kingdom
| | - Ana Paula C. A. Lima
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Maruyama SR, Fuzo CA, Oliveira AER, Rogerio LA, Takamiya NT, Pessenda G, de Melo EV, da Silva AM, Jesus AR, Carregaro V, Nakaya HI, Almeida RP, da Silva JS. Insight Into the Long Noncoding RNA and mRNA Coexpression Profile in the Human Blood Transcriptome Upon Leishmania infantum Infection. Front Immunol 2022; 13:784463. [PMID: 35370994 PMCID: PMC8965071 DOI: 10.3389/fimmu.2022.784463] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 02/11/2022] [Indexed: 12/13/2022] Open
Abstract
Visceral leishmaniasis (VL) is a vector-borne infectious disease that can be potentially fatal if left untreated. In Brazil, it is caused by Leishmania infantum parasites. Blood transcriptomics allows us to assess the molecular mechanisms involved in the immunopathological processes of several clinical conditions, namely, parasitic diseases. Here, we performed mRNA sequencing of peripheral blood from patients with visceral leishmaniasis during the active phase of the disease and six months after successful treatment, when the patients were considered clinically cured. To strengthen the study, the RNA-seq data analysis included two other non-diseased groups composed of healthy uninfected volunteers and asymptomatic individuals. We identified thousands of differentially expressed genes between VL patients and non-diseased groups. Overall, pathway analysis corroborated the importance of signaling involving interferons, chemokines, Toll-like receptors and the neutrophil response. Cellular deconvolution of gene expression profiles was able to discriminate cellular subtypes, highlighting the contribution of plasma cells and NK cells in the course of the disease. Beyond the biological processes involved in the immunopathology of VL revealed by the expression of protein coding genes (PCGs), we observed a significant participation of long noncoding RNAs (lncRNAs) in our blood transcriptome dataset. Genome-wide analysis of lncRNAs expression in VL has never been performed. lncRNAs have been considered key regulators of disease progression, mainly in cancers; however, their pattern regulation may also help to understand the complexity and heterogeneity of host immune responses elicited by L. infantum infections in humans. Among our findings, we identified lncRNAs such as IL21-AS1, MIR4435-2HG and LINC01501 and coexpressed lncRNA/mRNA pairs such as CA3-AS1/CA1, GASAL1/IFNG and LINC01127/IL1R1-IL1R2. Thus, for the first time, we present an integrated analysis of PCGs and lncRNAs by exploring the lncRNA–mRNA coexpression profile of VL to provide insights into the regulatory gene network involved in the development of this inflammatory and infectious disease.
Collapse
Affiliation(s)
- Sandra Regina Maruyama
- Department of Genetics and Evolution, Center for Biological Sciences and Health, Federal University of São Carlos, São Carlos, Brazil
| | - Carlos Alessandro Fuzo
- Department of Clinical Analyses, Toxicology and Food Sciences, Ribeirão Preto School of Pharmaceutics Sciences, University of São Paulo, Ribeirão Preto, Brazil
| | - Antonio Edson R Oliveira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luana Aparecida Rogerio
- Department of Genetics and Evolution, Center for Biological Sciences and Health, Federal University of São Carlos, São Carlos, Brazil
| | - Nayore Tamie Takamiya
- Department of Genetics and Evolution, Center for Biological Sciences and Health, Federal University of São Carlos, São Carlos, Brazil
| | - Gabriela Pessenda
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Enaldo Vieira de Melo
- Department of Medicine, University Hospital-Empresa Brasileira de Serviços Hospitalares (EBSERH), Federal University of Sergipe, Aracaju, Brazil
| | - Angela Maria da Silva
- Department of Medicine, University Hospital-Empresa Brasileira de Serviços Hospitalares (EBSERH), Federal University of Sergipe, Aracaju, Brazil
| | - Amélia Ribeiro Jesus
- Department of Medicine, University Hospital-Empresa Brasileira de Serviços Hospitalares (EBSERH), Federal University of Sergipe, Aracaju, Brazil
| | - Vanessa Carregaro
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Roque Pacheco Almeida
- Department of Medicine, University Hospital-Empresa Brasileira de Serviços Hospitalares (EBSERH), Federal University of Sergipe, Aracaju, Brazil
| | - João Santana da Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Fiocruz-Bi-Institutional Translational Medicine Platform, Ribeirão Preto, Brazil
| |
Collapse
|
13
|
Gardinassi LG, Maruyama SR, Cantacessi C. Editorial: Systems Biology of Hosts, Parasites and Vectors. Front Cell Infect Microbiol 2021; 11:796475. [PMID: 34805003 PMCID: PMC8599433 DOI: 10.3389/fcimb.2021.796475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 10/20/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Luiz Gustavo Gardinassi
- Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Brazil
| | - Sandra Regina Maruyama
- Department of Genetics and Evolution, Center for Biological Sciences and Health, Federal University of São Carlos, São Carlos, Brazil
| | - Cinzia Cantacessi
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
14
|
Panditrao G, Ganguli P, Sarkar RR. Delineating infection strategies of Leishmania donovani secretory proteins in Human through host-pathogen protein Interactome prediction. Pathog Dis 2021; 79:6408463. [PMID: 34677584 DOI: 10.1093/femspd/ftab051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 10/20/2021] [Indexed: 12/11/2022] Open
Abstract
Interactions of Leishmania donovani secretory virulence factors with the host proteins and their interplay during the infection process in humans is poorly studied in Visceral Leishmaniasis. Lack of a holistic study of pathway level de-regulations caused due to these virulence factors leads to a poor understanding of the parasite strategies to subvert the host immune responses, secure its survival inside the host and further the spread of infection to the visceral organs. In this study, we propose a computational workflow to predict host-pathogen protein interactome of L.donovani secretory virulence factors with human proteins combining sequence-based Interolog mapping and structure-based Domain Interaction mapping techniques. We further employ graph theoretical approaches and shortest path methods to analyze the interactome. Our study deciphers the infection paths involving some unique and understudied disease-associated signaling pathways influencing the cellular phenotypic responses in the host. Our statistical analysis based in silico knockout study unveils for the first time UBC, 1433Z and HS90A mediator proteins as potential immunomodulatory candidates through which the virulence factors employ the infection paths. These identified pathways and novel mediator proteins can be effectively used as possible targets to control and modulate the infection process further aiding in the treatment of Visceral Leishmaniasis.
Collapse
Affiliation(s)
- Gauri Panditrao
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune 411008, Maharashtra, India
| | - Piyali Ganguli
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune 411008, Maharashtra, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Ram Rup Sarkar
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune 411008, Maharashtra, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| |
Collapse
|
15
|
Farias Amorim C, O. Novais F, Nguyen BT, Nascimento MT, Lago J, Lago AS, Carvalho LP, Beiting DP, Scott P. Localized skin inflammation during cutaneous leishmaniasis drives a chronic, systemic IFN-γ signature. PLoS Negl Trop Dis 2021; 15:e0009321. [PMID: 33793565 PMCID: PMC8043375 DOI: 10.1371/journal.pntd.0009321] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 04/13/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
Cutaneous leishmaniasis is a localized infection controlled by CD4+ T cells that produce IFN-γ within lesions. Phagocytic cells recruited to lesions, such as monocytes, are then exposed to IFN-γ which triggers their ability to kill the intracellular parasites. Consistent with this, transcriptional analysis of patient lesions identified an interferon stimulated gene (ISG) signature. To determine whether localized L. braziliensis infection triggers a systemic immune response that may influence the disease, we performed RNA sequencing (RNA-seq) on the blood of L. braziliensis-infected patients and healthy controls. Functional enrichment analysis identified an ISG signature as the dominant transcriptional response in the blood of patients. This ISG signature was associated with an increase in monocyte- and macrophage-specific marker genes in the blood and elevated serum levels IFN-γ. A cytotoxicity signature, which is a dominant feature in the lesions, was also observed in the blood and correlated with an increased abundance of cytolytic cells. Thus, two transcriptional signatures present in lesions were found systemically, although with a substantially reduced number of differentially expressed genes (DEGs). Finally, we found that the number of DEGs and ISGs in leishmaniasis was similar to tuberculosis-another localized infection-but significantly less than observed in malaria. In contrast, the cytolytic signature and increased cytolytic cell abundance was not found in tuberculosis or malaria. Our results indicate that systemic signatures can reflect what is occurring in leishmanial lesions. Furthermore, the presence of an ISG signature in blood monocytes and macrophages suggests a mechanism to limit systemic spread of the parasite, as well as enhance parasite control by pre-activating cells prior to lesion entry.
Collapse
Affiliation(s)
- Camila Farias Amorim
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Fernanda O. Novais
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Ba T. Nguyen
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Mauricio T. Nascimento
- Serviço de Imunologia, Complexo Hospitalar Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Laboratório de Pesquisas Clínicas do Instituto de Pesquisas Gonçalo Moniz–Fiocruz, Salvador, Brazil
| | - Jamile Lago
- Serviço de Imunologia, Complexo Hospitalar Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Laboratório de Pesquisas Clínicas do Instituto de Pesquisas Gonçalo Moniz–Fiocruz, Salvador, Brazil
| | - Alexsandro S. Lago
- Serviço de Imunologia, Complexo Hospitalar Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Laboratório de Pesquisas Clínicas do Instituto de Pesquisas Gonçalo Moniz–Fiocruz, Salvador, Brazil
| | - Lucas P. Carvalho
- Serviço de Imunologia, Complexo Hospitalar Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
- Laboratório de Pesquisas Clínicas do Instituto de Pesquisas Gonçalo Moniz–Fiocruz, Salvador, Brazil
| | - Daniel P. Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, United States of America
| |
Collapse
|
16
|
Should an intersection between visceral leishmaniasis endemicity and the COVID-19 pandemic be considered? Med Hypotheses 2020; 144:110289. [PMID: 33254591 PMCID: PMC7501079 DOI: 10.1016/j.mehy.2020.110289] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022]
Abstract
The COVID-19 pandemic caused by the infection with the novel Coronavirus SARS-CoV-2, revealed individual and global vulnerabilities, in which we highlight the social, economic, and political aspects and the health systems’ organization in the countries. Brazil remains with a high transmission rate and presents a centripetal distribution as observed through a more sustained growth in the number of municipalities affected, outlining a profile of invasion of poor communities. Several vulnerabilities overlap with precarious housing conditions, lack of basic sanitation, malnutrition, and endemicity for neglected chronic diseases such as visceral leishmaniasis (VL). COVID-19 and VL evidently do not share clinical features, but exactly because of the distinct immunopathogenesis between the diseases, patients with VL may present a vulnerability in the immune system against antiviral responses. Considering that VL susceptibility seems to be related to an inefficient and polarized immune response, it is likely that in endemic areas, the overlap of social weaknesses added to individual vulnerability by immune polarization may aggravate the COVID-19 condition. In this sense, we reinforce that possible relationships between endemic neglected diseases such as VL and pandemic SARS-CoV-2 infection need to be further considered and investigated.
Collapse
|
17
|
Kumar A, Pandey SC, Samant M. DNA-based microarray studies in visceral leishmaniasis: identification of biomarkers for diagnostic, prognostic and drug target for treatment. Acta Trop 2020; 208:105512. [PMID: 32389452 DOI: 10.1016/j.actatropica.2020.105512] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 03/04/2020] [Accepted: 04/18/2020] [Indexed: 02/05/2023]
Abstract
Visceral leishmaniasis (VL) is one of the major infectious diseases affecting the poorest regions of the world. Current therapy is not very much satisfactory. The alarming rise of drug resistance and the unavailability of an effective vaccine against VL urges research towards identifying new targets or biomarkers for its effective treatment. New technology developments offer some fresh hope in its diagnosis, treatment, and control. DNA microarray approach is now broadly used in parasitology research to facilitate the thoughtful of mechanisms of disease and identification of drug targets and biomarkers for diagnostic and therapeutic development. An electronic search on "VL" and "Microarray" was conducted in Medline and Scopus and papers published in the English mentioning use of DNA microarray on VL were selected and read to write this paper review. Functional analysis and interpretation of microarray results remain very challenging due to the inherent nature of experimental workflows, access, cost, and complexity of data obtained. We have explained and emphasized the use of curate knowledge of microarray in the case of VL for the identification of therapeutic target and biomarker and their selection/implementation in clinical use.
Collapse
Affiliation(s)
- Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur (Chhattisgarh), INDIA
| | - Satish Chandra Pandey
- Cell and Molecular biology laboratory, Department of Zoology, Kumaun University, SSJ Campus, Almora (Uttarakhand), INDIA; Department of Biotechnology, Kumaun University Nainital, Bhimtal Campus, Bhimtal, Nainital (Uttarakhand), INDIA
| | - Mukesh Samant
- Cell and Molecular biology laboratory, Department of Zoology, Kumaun University, SSJ Campus, Almora (Uttarakhand), INDIA.
| |
Collapse
|
18
|
Gardinassi LG, Souza COS, Sales-Campos H, Fonseca SG. Immune and Metabolic Signatures of COVID-19 Revealed by Transcriptomics Data Reuse. Front Immunol 2020; 11:1636. [PMID: 32670298 PMCID: PMC7332781 DOI: 10.3389/fimmu.2020.01636] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/18/2020] [Indexed: 12/21/2022] Open
Abstract
The current pandemic of coronavirus disease 19 (COVID-19) has affected millions of individuals and caused thousands of deaths worldwide. The pathophysiology of the disease is complex and mostly unknown. Therefore, identifying the molecular mechanisms that promote progression of the disease is critical to overcome this pandemic. To address such issues, recent studies have reported transcriptomic profiles of cells, tissues and fluids from COVID-19 patients that mainly demonstrated activation of humoral immunity, dysregulated type I and III interferon expression, intense innate immune responses and inflammatory signaling. Here, we provide novel perspectives on the pathophysiology of COVID-19 using robust functional approaches to analyze public transcriptome datasets. In addition, we compared the transcriptional signature of COVID-19 patients with individuals infected with SARS-CoV-1 and Influenza A (IAV) viruses. We identified a core transcriptional signature induced by the respiratory viruses in peripheral leukocytes, whereas the absence of significant type I interferon/antiviral responses characterized SARS-CoV-2 infection. We also identified the higher expression of genes involved in metabolic pathways including heme biosynthesis, oxidative phosphorylation and tryptophan metabolism. A BTM-driven meta-analysis of bronchoalveolar lavage fluid (BALF) from COVID-19 patients showed significant enrichment for neutrophils and chemokines, which were also significant in data from lung tissue of one deceased COVID-19 patient. Importantly, our results indicate higher expression of genes related to oxidative phosphorylation both in peripheral mononuclear leukocytes and BALF, suggesting a critical role for mitochondrial activity during SARS-CoV-2 infection. Collectively, these data point for immunopathological features and targets that can be therapeutically exploited to control COVID-19.
Collapse
Affiliation(s)
- Luiz G. Gardinassi
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| | - Camila O. S. Souza
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Helioswilton Sales-Campos
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| | - Simone G. Fonseca
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| |
Collapse
|
19
|
Van Bockstal L, Bulté D, Van den Kerkhof M, Dirkx L, Mabille D, Hendrickx S, Delputte P, Maes L, Caljon G. Interferon Alpha Favors Macrophage Infection by Visceral Leishmania Species Through Upregulation of Sialoadhesin Expression. Front Immunol 2020; 11:1113. [PMID: 32582193 PMCID: PMC7296180 DOI: 10.3389/fimmu.2020.01113] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 05/07/2020] [Indexed: 12/24/2022] Open
Abstract
Type I interferons (IFNs) induced by an endogenous Leishmania RNA virus or exogenous viral infections have been shown to exacerbate infections with New World Cutaneous Leishmania parasites, however, the impact of type I IFNs in visceral Leishmania infections and implicated mechanisms remain to be unraveled. This study assessed the impact of type I IFN on macrophage infection with L. infantum and L. donovani and the implication of sialoadhesin (Siglec-1/CD169, Sn) as an IFN-inducible surface receptor. Stimulation of bone marrow-derived macrophages with type I IFN (IFN-α) significantly enhanced susceptibility to infection of reference laboratory strains and a set of recent clinical isolates. IFN-α particularly enhanced promastigote uptake. Enhanced macrophage susceptibility was linked to upregulated Sn surface expression as a major contributing factor to the infection exacerbating effect of IFN-α. Stimulation experiments in Sn-deficient macrophages, macrophage pretreatment with a monoclonal anti-Sn antibody or a novel bivalent anti-Sn nanobody and blocking of parasites with soluble Sn restored normal susceptibility levels. Infection of Sn-deficient mice with bioluminescent L. infantum promastigotes revealed a moderate, strain-dependent role for Sn during visceral infection under the used experimental conditions. These data indicate that IFN-responsive Sn expression can enhance the susceptibility of macrophages to infection with visceral Leishmania promastigotes and that targeting of Sn may have some protective effects in early infection.
Collapse
Affiliation(s)
- Lieselotte Van Bockstal
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Dimitri Bulté
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Magali Van den Kerkhof
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Laura Dirkx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Dorien Mabille
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Sarah Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Peter Delputte
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
20
|
Translational profiling of macrophages infected with Leishmania donovani identifies mTOR- and eIF4A-sensitive immune-related transcripts. PLoS Pathog 2020; 16:e1008291. [PMID: 32479529 PMCID: PMC7310862 DOI: 10.1371/journal.ppat.1008291] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 06/23/2020] [Accepted: 05/13/2020] [Indexed: 12/28/2022] Open
Abstract
The protozoan parasite Leishmania donovani (L. donovani) causes visceral leishmaniasis, a chronic infection which is fatal when untreated. Herein, we investigated whether in addition to altering transcription, L. donovani modulates host mRNA translation to establish a successful infection. Polysome-profiling revealed that one third of protein-coding mRNAs expressed in primary mouse macrophages are differentially translated upon infection with L. donovani promastigotes or amastigotes. Gene ontology analysis identified key biological processes enriched for translationally regulated mRNAs and were predicted to be either activated (e.g. chromatin remodeling and RNA metabolism) or inhibited (e.g. intracellular trafficking and antigen presentation) upon infection. Mechanistic in silico and biochemical analyses showed selective activation mTOR- and eIF4A-dependent mRNA translation, including transcripts encoding central regulators of mRNA turnover and inflammation (i.e. PABPC1, EIF2AK2, and TGF-β). L. donovani survival within macrophages was favored under mTOR inhibition but was dampened by pharmacological blockade of eIF4A. Overall, this study uncovers a vast yet selective reprogramming of the host cell translational landscape early during L. donovani infection, and suggests that some of these changes are involved in host defense mechanisms while others are part of parasite-driven survival strategies. Further in vitro and in vivo investigation will shed light on the contribution of mTOR- and eIF4A-dependent translational programs to the outcome of visceral leishmaniasis.
Collapse
|
21
|
Leishmaniasis immunopathology-impact on design and use of vaccines, diagnostics and drugs. Semin Immunopathol 2020; 42:247-264. [PMID: 32152715 DOI: 10.1007/s00281-020-00788-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/03/2020] [Indexed: 12/18/2022]
Abstract
Leishmaniasis is a disease complex caused by 20 species of protozoan parasites belonging to the genus Leishmania. In humans, it has two main clinical forms, visceral leishmaniasis (VL) and cutaneous or tegumentary leishmaniasis (CL), as well as several other cutaneous manifestations in a minority of cases. In the mammalian host Leishmania parasites infect different populations of macrophages where they multiply and survive in the phagolysosomal compartment. The progression of both VL and CL depends on the maintenance of a parasite-specific immunosuppressive state based around this host macrophage infection. The complexity and variation of immune responses and immunopathology in humans and the different host interactions of the different Leishmania species has an impact upon the effectiveness of vaccines, diagnostics and drugs.
Collapse
|
22
|
Adriaensen W, Cuypers B, Cordero CF, Mengasha B, Blesson S, Cnops L, Kaye PM, Alves F, Diro E, van Griensven J. Host transcriptomic signature as alternative test-of-cure in visceral leishmaniasis patients co-infected with HIV. EBioMedicine 2020; 55:102748. [PMID: 32361248 PMCID: PMC7195535 DOI: 10.1016/j.ebiom.2020.102748] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/03/2020] [Accepted: 03/19/2020] [Indexed: 12/14/2022] Open
Abstract
Background Visceral leishmaniasis (VL) treatment in HIV patients very often fails and is followed by high relapse and case-fatality rates. Hence, treatment efficacy assessment is imperative but based on invasive organ aspiration for parasite detection. In the search of a less-invasive alternative and because the host immune response is pivotal for treatment outcome in immunocompromised VL patients, we studied changes in the whole blood transcriptional profile of VL-HIV patients during treatment. Methods Embedded in a clinical trial in Northwest Ethiopia, RNA-Seq was performed on whole blood samples of 28 VL-HIV patients before and after completion of a 29-day treatment regimen of AmBisome or AmBisome/miltefosine. Pathway analyses were combined with a machine learning approach to establish a clinically-useful 4-gene set. Findings Distinct signatures of differentially expressed genes between D0 and D29 were identified for patients who failed treatment and were successfully treated. Pathway analyses in the latter highlighted a downregulation of genes associated with host cellular activity and immunity, and upregulation of antimicrobial peptide activity in phagolysosomes. No signs of disease remission nor pathway enrichment were observed in treatment failure patients. Next, we identified a 4-gene pre-post signature (PRSS33, IL10, SLFN14, HRH4) that could accurately discriminate treatment outcome at end of treatment (D29), displaying an average area-under-the-ROC-curve of 0.95 (CI: 0.75–1.00). Interpretation A simple blood-based signature thus holds significant promise to facilitate treatment efficacy monitoring and provide an alternative test-of-cure to guide patient management in VL-HIV patients. Funding Project funding was provided by the AfricoLeish project, supported by the European Union Seventh Framework Programme (EU FP7).
Collapse
Affiliation(s)
- Wim Adriaensen
- Unit of Neglected Tropical Diseases, Department of Clinical Sciences, Institute of Tropical Medicine, Nationalestraat 122, 2000 Antwerp, Belgium.
| | - Bart Cuypers
- Unit of Molecular Parasitology, Department of Biomedical Sciences, Institute of Tropical Medicine, Nationalestraat 122, 2000 Antwerp, Belgium; Adrem Data Lab, Department of Mathematics and Computer Science, University of Antwerp, Middelheim 1, 2020 Antwerp, Belgium
| | - Carlota F Cordero
- Unit of Neglected Tropical Diseases, Department of Clinical Sciences, Institute of Tropical Medicine, Nationalestraat 122, 2000 Antwerp, Belgium
| | - Bewketu Mengasha
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Séverine Blesson
- Drugs for Neglected Diseases initiative, 15 Chemin Louis-Dunant, 1202 Geneva, Switzerland
| | - Lieselotte Cnops
- Unit of Neglected Tropical Diseases, Department of Clinical Sciences, Institute of Tropical Medicine, Nationalestraat 122, 2000 Antwerp, Belgium
| | - Paul M Kaye
- York Biomedical Research Institute, University of York, United Kingdom
| | - Fabiana Alves
- Drugs for Neglected Diseases initiative, 15 Chemin Louis-Dunant, 1202 Geneva, Switzerland
| | - Ermias Diro
- Leishmaniasis Research and Treatment Centre, University of Gondar, Gondar, Ethiopia
| | - Johan van Griensven
- Unit of Neglected Tropical Diseases, Department of Clinical Sciences, Institute of Tropical Medicine, Nationalestraat 122, 2000 Antwerp, Belgium
| |
Collapse
|
23
|
Gatto M, Borim PA, Wolf IR, Fukuta da Cruz T, Ferreira Mota GA, Marques Braz AM, Casella Amorim B, Targino Valente G, de Assis Golim M, Venturini J, Araújo Junior JP, Pontillo A, Sartori A. Transcriptional analysis of THP-1 cells infected with Leishmania infantum indicates no activation of the inflammasome platform. PLoS Negl Trop Dis 2020; 14:e0007949. [PMID: 31961876 PMCID: PMC6994165 DOI: 10.1371/journal.pntd.0007949] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 01/31/2020] [Accepted: 11/25/2019] [Indexed: 12/31/2022] Open
Abstract
Leishmaniasis is caused by intracellular parasites transmitted to vertebrates by sandfly bites. Clinical manifestations include cutaneous, mucosal or visceral involvement depending upon the host immune response and the parasite species. To assure their survival inside macrophages, these parasites developed a plethora of highly successful strategies to manipulate various immune system pathways. Considering that inflammasome activation is critical for the establishment of a protective immune response in many parasite infections, in this study we determined the transcriptome of THP-1 cells after infection with L. infantum, with a particular focus on the inflammasome components. To this end, the human cell line THP-1, previously differentiated into macrophages by PMA treatment, was infected with L. infantum promastigotes. Differentiated THP-1 cells were also stimulated with LPS to be used as a comparative parameter. The gene expression signature was determined 8 hours after by RNA-seq technique. Infected or uninfected THP-1 cells were stimulated with nigericin (NIG) to measure active caspase-1 and TNF-α, IL-6 and IL-1β levels in culture supernatants after 8, 24 and 48 hours. L. infantum triggered a gene expression pattern more similar to non-infected THP-1 cells and very distinct from LPS-stimulated cells. Some of the most up-regulated genes in L. infantum-infected cells were CDC20, CSF1, RPS6KA1, CD36, DUSP2, DUSP5, DUSP7 and TNFAIP3. Some up-regulated GO terms in infected cells included cell coagulation, regulation of MAPK cascade, response to peptide hormone stimulus, negative regulation of transcription from RNA polymerase II promoter and nerve growth factor receptor signaling pathway. Infection was not able to induce the expression of genes associated with the inflammasome signaling pathway. This finding was confirmed by the absence of caspase-1 activation and IL-1β production after 8, 24 and 48 hours of infection. Our results indicate that L. infantum was unable to activate the inflammasomes during the initial interaction with THP-1 cells. Visceral leishmaniasis, caused by Leishmania infantum, is a disease that affects millions of people worldwide. The entry of microorganisms into the host is commonly associated with activation of a multiprotein platform called inflammasome whose assembly culminates in caspase-1 activation and IL-1β production. ILβ activates other cells and effector mechanisms leading to clearance of pathogens. However, the involvement of inflammasomes in the human infection with L. infantum is poorly known. To investigate the parasite-host interaction is fundamental to understand the immunopathogenesis of visceral leishmaniasis and to allow the development of new therapeutic strategies. In this study, we used RNA-seq, a tool that allowed to investigate the global gene expression of THP-1 cells, which is a macrophage-like human cell line, infected with L. infantum. By using computational analysis, this approach allowed us to evaluate the expression of genes that compose the inflammasomes pathway and other gene networks and signaling pathways triggered after infection. This analysis indicated that, unlike species causing cutaneous leishmaniasis, L. infantum did not induce the expression of genes of inflammasome pathways, nor caspase-1 activation or IL-1β production, possibly reflecting a parasite strategy to manipulate immune system and therefore, to allow its survival inside the cells.
Collapse
Affiliation(s)
- Mariana Gatto
- Tropical Diseases Department, Botucatu Medical School – UNESP, Botucatu, Brazil
- * E-mail:
| | | | - Ivan Rodrigo Wolf
- Bioprocess and Biotechnology Department, Agronomic Sciences School – UNESP, Botucatu, Brazil
| | - Taís Fukuta da Cruz
- Microbiology and Immunology Department, Biosciences Institute - UNESP, Botucatu, Brazil
| | | | | | | | | | | | | | | | | | - Alexandrina Sartori
- Tropical Diseases Department, Botucatu Medical School – UNESP, Botucatu, Brazil
| |
Collapse
|
24
|
Forrester S, Siefert K, Ashwin H, Brown N, Zelmar A, James S, Lagos D, Timmis J, Chatterjee M, Mottram JC, Croft SL, Kaye PM. Tissue-specific transcriptomic changes associated with AmBisome® treatment of BALB/c mice with experimental visceral leishmaniasis. Wellcome Open Res 2019; 4:198. [PMID: 31976381 PMCID: PMC6961418 DOI: 10.12688/wellcomeopenres.15606.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2019] [Indexed: 01/11/2023] Open
Abstract
Background: Liposomal amphotericin B (AmBisome®) as a treatment modality for visceral leishmaniasis (VL) has had significant impact on patient care in some but not all regions where VL is endemic. As the mode of action of AmBisome® in vivo is poorly understood, we compared the tissue-specific transcriptome in drug-treated vs untreated mice with experimental VL. Methods: BALB/c mice infected with L. donovani were treated with 8mg/kg AmBisome®, resulting in parasite elimination from liver and spleen over a 7-day period. At day 1 and day 7 post treatment (R x+1 and R x+7), transcriptomic profiling was performed on spleen and liver tissue from treated and untreated mice and uninfected mice. BALB/c mice infected with M. bovis BCG (an organism resistant to amphotericin B) were analysed to distinguish between direct effects of AmBisome® and those secondary to parasite death. Results: AmBisome® treatment lead to rapid parasitological clearance. At R x+1, spleen and liver displayed only 46 and 88 differentially expressed (DE) genes (P<0.05; 2-fold change) respectively. In liver, significant enrichment was seen for pathways associated with TNF, fatty acids and sterol biosynthesis. At R x+7, the number of DE genes was increased (spleen, 113; liver 400). In spleen, these included many immune related genes known to be involved in anti-leishmanial immunity. In liver, changes in transcriptome were largely accounted for by loss of granulomas. PCA analysis indicated that treatment only partially restored homeostasis. Analysis of BCG-infected mice treated with AmBisome® revealed a pattern of immune modulation mainly targeting macrophage function. Conclusions: Our data indicate that the tissue response to AmBisome® treatment varies between target organs and that full restoration of homeostasis is not achieved at parasitological cure. The pathways required to restore homeostasis deserve fuller attention, to understand mechanisms associated with treatment failure and relapse and to promote more rapid restoration of immune competence.
Collapse
Affiliation(s)
- Sarah Forrester
- York Biomedical Research Institute, University of York, York, YO10 5DD, UK
| | - Karin Siefert
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Helen Ashwin
- York Biomedical Research Institute, University of York, York, YO10 5DD, UK
| | - Najmeeyah Brown
- York Biomedical Research Institute, University of York, York, YO10 5DD, UK
| | - Andrea Zelmar
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Sally James
- Biosciences Technology Facility, University of York, York, YO10 5DD, UK
| | - Dimitris Lagos
- York Biomedical Research Institute, University of York, York, YO10 5DD, UK
| | - Jon Timmis
- Department of Electronic Engineering, University of York, UK, York, YO10 5DD, UK
| | - Mitali Chatterjee
- Department of Pharmacology, Jawaharlal Institute of Post Graduate Medical Education and Research, Kolkata, 700 020, India
| | - Jeremy C. Mottram
- York Biomedical Research Institute, University of York, York, YO10 5DD, UK
| | - Simon L. Croft
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Paul M. Kaye
- York Biomedical Research Institute, University of York, York, YO10 5DD, UK
| |
Collapse
|
25
|
Agallou M, Athanasiou E, Kammona O, Tastsoglou S, Hatzigeorgiou AG, Kiparissides C, Karagouni E. Transcriptome Analysis Identifies Immune Markers Related to Visceral Leishmaniasis Establishment in the Experimental Model of BALB/c Mice. Front Immunol 2019; 10:2749. [PMID: 31849951 PMCID: PMC6902045 DOI: 10.3389/fimmu.2019.02749] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/11/2019] [Indexed: 12/29/2022] Open
Abstract
Visceral leishmaniasis (VL) caused by Leishmania donovani and L. infantum is a potentially fatal disease. To date there are no registered vaccines for disease prevention despite the fact that several vaccines are in preclinical development. Thus, new strategies are needed to improve vaccine efficacy based on a better understanding of the mechanisms mediating protective immunity and mechanisms of host immune responses subversion by immunopathogenic components of Leishmania. We found that mice vaccinated with CPA162−189-loaded p8-PLGA nanoparticles, an experimental nanovaccine, induced the differentiation of antigen-specific CD8+ T cells in spleen compared to control mice, characterized by increased dynamics of proliferation and high amounts of IFN-γ production after ex vivo re-stimulation with CPA162−189 antigen. Vaccination with CPA162−189-loaded p8-PLGA nanoparticles resulted in about 80% lower parasite load in spleen and liver at 4 weeks after challenge with L. infantum promastigotes as compared to control mice. However, 16 weeks after infection the parasite load in spleen was comparable in both mouse groups. Decreased protection levels in vaccinated mice were followed by up-regulation of the anti-inflammatory IL-10 production although at lower levels in comparison to control mice. Microarray analysis in spleen tissue at 4 weeks post challenge revealed different immune-related profiles among the two groups. Specifically, vaccinated mice were characterized by similar profile to naïve mice. On the other hand, the transcriptome of the non-vaccinated mice was dominated by increased expression of genes related to interferon type I, granulocyte chemotaxis, and immune cells suppression. This profile was significantly enriched at 16 weeks post challenge, a time-point which is relative to disease establishment, and was common for both groups, further suggesting that type I signaling and granulocyte influx has a significant role in disease establishment, pathogenesis and eventually in decreased vaccine efficacy for stimulating long-term protection. Overall, we put a spotlight on host immune networks during active VL as potential targets to improve and design more effective vaccines against disease.
Collapse
Affiliation(s)
- Maria Agallou
- Parasite Immunology Group, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Evita Athanasiou
- Parasite Immunology Group, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Olga Kammona
- Chemical Process & Energy Resources Institute, Centre for Research and Technology Hellas, Thessaloniki, Greece
| | - Spyros Tastsoglou
- DIANA-Lab, Department of Electrical & Computer Engineering, University of Thessaly, Volos, Greece
| | - Artemis G Hatzigeorgiou
- DIANA-Lab, Department of Electrical & Computer Engineering, University of Thessaly, Volos, Greece.,DIANA-Lab, Hellenic Pasteur Institute, Athens, Greece
| | - Costas Kiparissides
- Chemical Process & Energy Resources Institute, Centre for Research and Technology Hellas, Thessaloniki, Greece.,Department of Chemical Engineering, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evdokia Karagouni
- Parasite Immunology Group, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| |
Collapse
|
26
|
Fakiola M, Singh OP, Syn G, Singh T, Singh B, Chakravarty J, Sundar S, Blackwell JM. Transcriptional blood signatures for active and amphotericin B treated visceral leishmaniasis in India. PLoS Negl Trop Dis 2019; 13:e0007673. [PMID: 31419223 PMCID: PMC6713396 DOI: 10.1371/journal.pntd.0007673] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 08/28/2019] [Accepted: 07/30/2019] [Indexed: 12/13/2022] Open
Abstract
Amphotericin B provides improved therapy for visceral leishmaniasis (VL) caused by Leishmania donovani, with single dose liposomal-encapsulated Ambisome providing the best cure rates. The VL elimination program aims to reduce the incidence rate in the Indian subcontinent to <1/10,000 population/year. Ability to predict which asymptomatic individuals (e.g. anti-leishmanial IgG and/or Leishmania-specific modified Quantiferon positive) will progress to clinical VL would help in monitoring disease outbreaks. Here we examined whole blood transcriptional profiles associated with asymptomatic infection, active disease, and in treated cases. Two independent microarray experiments were performed, with analysis focussed primarily on differentially expressed genes (DEGs) concordant across both experiments. No DEGs were identified for IgG or Quantiferon positive asymptomatic groups compared to negative healthy endemic controls. We therefore concentrated on comparing concordant DEGs from active cases with all healthy controls, and in examining differences in the transcriptome following different regimens of drug treatment. In these comparisons 6 major themes emerged: (i) expression of genes and enrichment of gene sets associated with erythrocyte function in active cases; (ii) strong evidence for enrichment of gene sets involved in cell cycle in comparing active cases with healthy controls; (iii) identification of IFNG encoding interferon-γ as the major hub gene in concordant gene expression patterns across experiments comparing active cases with healthy controls or with treated cases; (iv) enrichment for interleukin signalling (IL-1/3/4/6/7/8) and a prominent role for CXCL10/9/11 and chemokine signalling pathways in comparing active cases with treated cases; (v) the novel identification of Aryl Hydrocarbon Receptor signalling as a significant canonical pathway when comparing active cases with healthy controls or with treated cases; and (vi) global expression profiling support for more effective cure at day 30 post-treatment with a single dose of liposomal encapsulated amphotericin B compared to multi-dose non-liposomal amphotericin B treatment over 30 days. (296 words; 300 words allowed).
Collapse
Affiliation(s)
- Michaela Fakiola
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- INGM-National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi" Milan, Milan, Italy
| | - Om Prakash Singh
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Genevieve Syn
- Telethon Kids Institute, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Toolika Singh
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Bhawana Singh
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Jaya Chakravarty
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Jenefer M. Blackwell
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Telethon Kids Institute, The University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
27
|
Evaluation of methods for detection of asymptomatic individuals infected with Leishmania infantum in the state of Piauí, Brazil. PLoS Negl Trop Dis 2019; 13:e0007493. [PMID: 31260440 PMCID: PMC6625727 DOI: 10.1371/journal.pntd.0007493] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 07/12/2019] [Accepted: 05/24/2019] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Visceral Leishmaniasis in humans presents with fever, anemia, and splenomegaly and can be lethal if not treated. Nevertheless, the majority of Leishmania infantum-infected individuals does not manifest symptoms and remain so provided they are not immunosuppressed. In this work, the performance of different tests was evaluated to detect asymptomatic individuals who were living in Teresina, Piauí state, Brazil, an endemic area for VL. METHODOLOGY L. infantum-specific antibodies were detected by ELISA and two different rapid immunochromatographic (IC) diagnostic tests, Kalazar Detect and OnSite, and parasitic loads were detected by real time PCR [qPCR]. Additionally, we measured levels of the biomarkers monokine induced by IFN-γ (MIG) and IFN-γ-induced protein 10 (IP-10) before and after stimulation of whole blood with soluble Leishmania antigen [SLA]. PRINCIPAL FINDINGS Kalazar Detect and OnSite detected, respectively, 76% and 64% of patients presenting with active Visceral Leishmaniasis; 50% and 57% of patients remained positive in these tests, respectively, after treatment. Of the healthy participants in the study who were living in the endemic area, only 1.7% were positive with both of the IC tests. On the other hand, reactivity in ELISA tests revealed that 13% of these individuals presented asymptomatic infections; among VL patients, 84% presenting with active disease were reactive in ELISA, and after treatment, 55.5% were seropositive. L. infantum DNA was present in the blood of 37.9% of infected individuals living in the endemic area, while IP-10 and MIG biomarkers were detected in 26.7% of them. The greatest concordance of positivity occurred between ELISA and qPCR. CONCLUSION The association of different techniques can detect asymptomatic infections, however, more research is necessary to develop ideal biomarkers that are simple to use in the clinic and in field studies in areas endemic for Visceral Leishmaniasis.
Collapse
|
28
|
Chauhan SB, Faleiro R, Kumar R, Ng S, Singh B, Singh OP, Singh SS, Amante F, Rivera FDL, Rai M, Chakravarty J, Sacks D, Nylen S, Sundar S, Engwerda C. Interleukin 2 is an Upstream Regulator of CD4+ T Cells From Visceral Leishmaniasis Patients With Therapeutic Potential. J Infect Dis 2019; 220:163-173. [PMID: 30796820 PMCID: PMC6775044 DOI: 10.1093/infdis/jiz074] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 02/19/2019] [Indexed: 12/14/2022] Open
Abstract
Control of visceral leishmaniasis (VL) caused by Leishmania donovani requires interferon-γ production by CD4+ T cells. In VL patients, antiparasitic CD4+ T-cell responses are ineffective for unknown reasons. In this study, we measured the expression of genes associated with various immune functions in these cells from VL patients and compared them to CD4+ T cells from the same patients after drug treatment and from endemic controls. We found reduced GATA3, RORC, and FOXP3 gene expression in CD4+ T cells of VL patients, associated with reduced Th2, Th17, and FOXP3+CD4+ T regulatory cell frequencies in VL patient blood. Interleukin 2 (IL-2) was an important upstream regulator of CD4+ T cells from VL patients, and functional studies demonstrated the therapeutic potential of IL-2 for improving antiparasitic immunity. Together, these results provide new insights into the characteristics of CD4+ T cells from VL patients that can be used to improve antiparasitic immune responses.
Collapse
Affiliation(s)
- Shashi Bhushan Chauhan
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Praadesh, India
| | - Rebecca Faleiro
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Rajiv Kumar
- Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Susanna Ng
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Bhawana Singh
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Praadesh, India
| | - Om Prakash Singh
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Praadesh, India
| | - Siddharth Sankar Singh
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Praadesh, India
| | - Fiona Amante
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - Madhukar Rai
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Praadesh, India
| | - Jaya Chakravarty
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Praadesh, India
| | - David Sacks
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Susanne Nylen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Praadesh, India
| | | |
Collapse
|
29
|
Ashwin H, Seifert K, Forrester S, Brown N, MacDonald S, James S, Lagos D, Timmis J, Mottram JC, Croft SL, Kaye PM. Tissue and host species-specific transcriptional changes in models of experimental visceral leishmaniasis. Wellcome Open Res 2019; 3:135. [PMID: 30542664 PMCID: PMC6248268 DOI: 10.12688/wellcomeopenres.14867.2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2018] [Indexed: 12/19/2022] Open
Abstract
Background: Human visceral leishmaniasis, caused by infection with Leishmania donovani or L. infantum, is a potentially fatal disease affecting 50,000-90,000 people yearly in 75 disease endemic countries, with more than 20,000 deaths reported. Experimental models of infection play a major role in understanding parasite biology, host-pathogen interaction, disease pathogenesis, and parasite transmission. In addition, they have an essential role in the identification and pre-clinical evaluation of new drugs and vaccines. However, our understanding of these models remains fragmentary. Although the immune response to Leishmania donovani infection in mice has been extensively characterized, transcriptomic analysis capturing the tissue-specific evolution of disease has yet to be reported. Methods: We provide an analysis of the transcriptome of spleen, liver and peripheral blood of BALB/c mice infected with L. donovani. Where possible, we compare our data in murine experimental visceral leishmaniasis with transcriptomic data in the public domain obtained from the study of L. donovani-infected hamsters and patients with human visceral leishmaniasis. Digitised whole slide images showing the histopathology in spleen and liver are made available via a dedicated website, www.leishpathnet.org. Results: Our analysis confirms marked tissue-specific alterations in the transcriptome of infected mice over time and identifies previously unrecognized parallels and differences between murine, hamster and human responses to infection. We show commonality of interferon-regulated genes whilst confirming a greater activation of type 2 immune pathways in infected hamsters compared to mice. Cytokine genes and genes encoding immune checkpoints were markedly tissue specific and dynamic in their expression, and pathways focused on non-immune cells reflected tissue specific immunopathology. Our data also addresses the value of measuring peripheral blood transcriptomics as a potential window into underlying systemic disease. Conclusions: Our transcriptomic data, coupled with histopathologic analysis of the tissue response, provide an additional resource to underpin future mechanistic studies and to guide clinical research.
Collapse
Affiliation(s)
- Helen Ashwin
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Karin Seifert
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Sarah Forrester
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Najmeeyah Brown
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Sandy MacDonald
- Bioscience Technology Facility, Deptartment of Biology, University of York, York, YO10 5DD, UK
| | - Sally James
- Bioscience Technology Facility, Deptartment of Biology, University of York, York, YO10 5DD, UK
| | - Dimitris Lagos
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Jon Timmis
- Dept of Electronic Engineering, University of York, York, YO10 5DD, UK
| | - Jeremy C Mottram
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Simon L. Croft
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Paul M. Kaye
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| |
Collapse
|
30
|
Ashwin H, Seifert K, Forrester S, Brown N, MacDonald S, James S, Lagos D, Timmis J, Mottram JC, Croft SL, Kaye PM. Tissue and host species-specific transcriptional changes in models of experimental visceral leishmaniasis. Wellcome Open Res 2018; 3:135. [PMID: 30542664 PMCID: PMC6248268 DOI: 10.12688/wellcomeopenres.14867.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2018] [Indexed: 11/08/2023] Open
Abstract
Background: Human visceral leishmaniasis, caused by infection with Leishmania donovani or L. infantum, is a potentially fatal disease affecting 50,000-90,000 people yearly in 75 disease endemic countries, with more than 20,000 deaths reported. Experimental models of infection play a major role in understanding parasite biology, host-pathogen interaction, disease pathogenesis, and parasite transmission. In addition, they have an essential role in the identification and pre-clinical evaluation of new drugs and vaccines. However, our understanding of these models remains fragmentary. Although the immune response to Leishmania donovani infection in mice has been extensively characterized, transcriptomic analysis capturing the tissue-specific evolution of disease has yet to be reported. Methods: We provide an analysis of the transcriptome of spleen, liver and peripheral blood of BALB/c mice infected with L. donovani. Where possible, we compare our data in murine experimental visceral leishmaniasis with transcriptomic data in the public domain obtained from the study of L. donovani-infected hamsters and patients with human visceral leishmaniasis. Digitised whole slide images showing the histopathology in spleen and liver are made available via a dedicated website, www.leishpathnet.org. Results: Our analysis confirms marked tissue-specific alterations in the transcriptome of infected mice over time and identifies previously unrecognized parallels and differences between murine, hamster and human responses to infection. We show commonality of interferon-regulated genes whilst confirming a greater activation of type 2 immune pathways in infected hamsters compared to mice. Cytokine genes and genes encoding immune checkpoints were markedly tissue specific and dynamic in their expression, and pathways focused on non-immune cells reflected tissue specific immunopathology. Our data also addresses the value of measuring peripheral blood transcriptomics as a potential window into underlying systemic disease. Conclusions: Our transcriptomic data, coupled with histopathologic analysis of the tissue response, provide an additional resource to underpin future mechanistic studies and to guide clinical research.
Collapse
Affiliation(s)
- Helen Ashwin
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Karin Seifert
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Sarah Forrester
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Najmeeyah Brown
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Sandy MacDonald
- Bioscience Technology Facility, Deptartment of Biology, University of York, York, YO10 5DD, UK
| | - Sally James
- Bioscience Technology Facility, Deptartment of Biology, University of York, York, YO10 5DD, UK
| | - Dimitris Lagos
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Jon Timmis
- Dept of Electronic Engineering, University of York, York, YO10 5DD, UK
| | - Jeremy C Mottram
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| | - Simon L. Croft
- Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Paul M. Kaye
- Centre for Immunology and Infection, University of York, York, YO10 5DD, UK
| |
Collapse
|
31
|
Veras PST, Ramos PIP, de Menezes JPB. In Search of Biomarkers for Pathogenesis and Control of Leishmaniasis by Global Analyses of Leishmania-Infected Macrophages. Front Cell Infect Microbiol 2018; 8:326. [PMID: 30283744 PMCID: PMC6157484 DOI: 10.3389/fcimb.2018.00326] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/27/2018] [Indexed: 12/12/2022] Open
Abstract
Leishmaniasis is a vector-borne, neglected tropical disease with a worldwide distribution that can present in a variety of clinical forms, depending on the parasite species and host genetic background. The pathogenesis of this disease remains far from being elucidated because the involvement of a complex immune response orchestrated by host cells significantly affects the clinical outcome. Among these cells, macrophages are the main host cells, produce cytokines and chemokines, thereby triggering events that contribute to the mediation of the host immune response and, subsequently, to the establishment of infection or, alternatively, disease control. There has been relatively limited commercial interest in developing new pharmaceutical compounds to treat leishmaniasis. Moreover, advances in the understanding of the underlying biology of Leishmania spp. have not translated into the development of effective new chemotherapeutic compounds. As a result, biomarkers as surrogate disease endpoints present several potential advantages to be used in the identification of targets capable of facilitating therapeutic interventions considered to ameliorate disease outcome. More recently, large-scale genomic and proteomic analyses have allowed the identification and characterization of the pathways involved in the infection process in both parasites and the host, and these analyses have been shown to be more effective than studying individual molecules to elucidate disease pathogenesis. RNA-seq and proteomics are large-scale approaches that characterize genes or proteins in a given cell line, tissue, or organism to provide a global and more integrated view of the myriad biological processes that occur within a cell than focusing on an individual gene or protein. Bioinformatics provides us with the means to computationally analyze and integrate the large volumes of data generated by high-throughput sequencing approaches. The integration of genomic expression and proteomic data offers a rich multi-dimensional analysis, despite the inherent technical and statistical challenges. We propose that these types of global analyses facilitate the identification, among a large number of genes and proteins, those that hold potential as biomarkers. The present review focuses on large-scale studies that have identified and evaluated relevant biomarkers in macrophages in response to Leishmania infection.
Collapse
Affiliation(s)
- Patricia Sampaio Tavares Veras
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador, Brazil.,National Institute of Tropical Disease, Brasilia, Brazil
| | - Pablo Ivan Pereira Ramos
- Center for Data and Knowledge Integration for Health, Gonçalo Moniz Institute, Fiocruz-Bahia, Salvador, Brazil
| | | |
Collapse
|
32
|
Chambers DM, Moretti L, Zhang JJ, Cooper SW, Chambers DM, Santangelo PJ, Barker TH. LEM domain-containing protein 3 antagonizes TGFβ-SMAD2/3 signaling in a stiffness-dependent manner in both the nucleus and cytosol. J Biol Chem 2018; 293:15867-15886. [PMID: 30108174 DOI: 10.1074/jbc.ra118.003658] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/31/2018] [Indexed: 12/28/2022] Open
Abstract
Transforming growth factor-β (TGFβ) signaling through SMAD2/3 is an important driver of pathological fibrosis in multiple organ systems. TGFβ signaling and extracellular matrix (ECM) stiffness form an unvirtuous pathological circuit in which matrix stiffness drives activation of latent TGFβ, and TGFβ signaling then drives cellular stress and ECM synthesis. Moreover, ECM stiffness also appears to sensitize cells to exogenously activated TGFβ through unknown mechanisms. Here, using human fibroblasts, we explored the effect of ECM stiffness on a putative inner nuclear membrane protein, LEM domain-containing protein 3 (LEMD3), which is physically connected to the cell's actin cytoskeleton and inhibits TGFβ signaling. We showed that LEMD3-SMAD2/3 interactions are inversely correlated with ECM stiffness and TGFβ-driven luciferase activity and that LEMD3 expression is correlated with the mechanical response of the TGFβ-driven luciferase reporter. We found that actin polymerization but not cellular stress or LEMD3-nuclear-cytoplasmic couplings were necessary for LEMD3-SMAD2/3 interactions. Intriguingly, LEMD3 and SMAD2/3 frequently interacted in the cytosol, and we discovered LEMD3 was proteolytically cleaved into protein fragments. We confirmed that a consensus C-terminal LEMD3 fragment binds SMAD2/3 in a stiffness-dependent manner throughout the cell and is sufficient for antagonizing SMAD2/3 signaling. Using human lung biopsies, we observed that these nuclear and cytosolic interactions are also present in tissue and found that fibrotic tissues exhibit locally diminished and cytoplasmically shifted LEMD3-SMAD2/3 interactions, as noted in vitro Our work reveals novel LEMD3 biology and stiffness-dependent regulation of TGFβ by LEMD3, providing a novel target to antagonize pathological TGFβ signaling.
Collapse
Affiliation(s)
- Dwight M Chambers
- From the Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332
| | - Leandro Moretti
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, and
| | - Jennifer J Zhang
- From the Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332
| | - Spencer W Cooper
- From the Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332
| | - Davis M Chambers
- the College of Arts and Sciences, Georgia State University, Atlanta, Georgia 30303
| | - Philip J Santangelo
- From the Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia 30332
| | - Thomas H Barker
- Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22908, and
| |
Collapse
|
33
|
Leonor Fernandes Saraiva JP, Zubiria-Barrera C, Klassert TE, Lautenbach MJ, Blaess M, Claus RA, Slevogt H, König R. Combination of Classifiers Identifies Fungal-Specific Activation of Lysosome Genes in Human Monocytes. Front Microbiol 2017; 8:2366. [PMID: 29238336 PMCID: PMC5712586 DOI: 10.3389/fmicb.2017.02366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 11/16/2017] [Indexed: 12/31/2022] Open
Abstract
Blood stream infections can be caused by several pathogens such as viruses, fungi and bacteria and can cause severe clinical complications including sepsis. Delivery of appropriate and quick treatment is mandatory. However, it requires a rapid identification of the invading pathogen. The current gold standard for pathogen identification relies on blood cultures and these methods require a long time to gain the needed diagnosis. The use of in situ experiments attempts to identify pathogen specific immune responses but these often lead to heterogeneous biomarkers due to the high variability in methods and materials used. Using gene expression profiles for machine learning is a developing approach to discriminate between types of infection, but also shows a high degree of inconsistency. To produce consistent gene signatures, capable of discriminating fungal from bacterial infection, we have employed Support Vector Machines (SVMs) based on Mixed Integer Linear Programming (MILP). Combining classifiers by joint optimization constraining them to the same set of discriminating features increased the consistency of our biomarker list independently of leukocyte-type or experimental setup. Our gene signature showed an enrichment of genes of the lysosome pathway which was not uncovered by the use of independent classifiers. Moreover, our results suggest that the lysosome genes are specifically induced in monocytes. Real time qPCR of the identified lysosome-related genes confirmed the distinct gene expression increase in monocytes during fungal infections. Concluding, our combined classifier approach presented increased consistency and was able to "unmask" signaling pathways of less-present immune cells in the used datasets.
Collapse
Affiliation(s)
- João P Leonor Fernandes Saraiva
- Network Modeling, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany.,Integrated Research and Treatment Center, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | | | | | | | - Markus Blaess
- Integrated Research and Treatment Center, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Ralf A Claus
- Integrated Research and Treatment Center, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Hortense Slevogt
- Septomics Research Centre, Jena University Hospital, Jena, Germany
| | - Rainer König
- Network Modeling, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany.,Integrated Research and Treatment Center, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| |
Collapse
|
34
|
Quintela-Carvalho G, Luz NF, Celes FS, Zanette DL, Andrade D, Menezes D, Tavares NM, Brodskyn CI, Prates DB, Gonçalves MS, de Oliveira CI, Almeida RP, Bozza MT, Andrade BB, Borges VM. Heme Drives Oxidative Stress-Associated Cell Death in Human Neutrophils Infected with Leishmania infantum. Front Immunol 2017; 8:1620. [PMID: 29218050 PMCID: PMC5703736 DOI: 10.3389/fimmu.2017.01620] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/08/2017] [Indexed: 11/25/2022] Open
Abstract
Free heme is an inflammatory molecule capable of inducing migration and activation of neutrophils. Here, we examine the heme-driven oxidative stress-associated cell death mechanisms in human neutrophils infected with Leishmania infantum, an etiologic agent of visceral leishmaniasis (VL). We first performed exploratory analyses in a population of well characterized treatment-naïve VL patients as well as uninfected controls, who were part of previously reported studies. We noted a positive correlation between serum concentrations of heme with heme oxygenase-1 (HO-1) and lactate deydrogenase, as well as, a negative correlation between heme values and peripheral blood neutrophils counts. Moreover, in vitro infection with L. infantum in the presence of heme enhanced parasite burden in neutrophils, while increasing the production of reactive oxygen species and release of neutrophilic enzymes. Additional experiments demonstrated that treatment of infected neutrophils with ferrous iron (Fe+2), a key component of the heme molecule, resulted in increased parasite survival without affecting neutrophil activation status. Furthermore, stimulation of infected neutrophils with heme triggered substantial increases in HO-1 mRNA expression as well as in superoxide dismutase-1 enzymatic activity. Heme, but not Fe+2, induced oxidative stress-associated cell death. These findings indicate that heme promotes intracellular L. infantum survival via activation of neutrophil function and oxidative stress. This study opens new perspectives for the understanding of immunopathogenic mechanisms involving neutrophils in VL.
Collapse
Affiliation(s)
- Graziele Quintela-Carvalho
- Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil.,Instituto Federal de Educação, Ciência e Tecnologia Baiano (IFBaiano), Santa Inês, Brazil
| | - Nívea F Luz
- Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
| | - Fabiana S Celes
- Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Dalila L Zanette
- Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
| | - Daniela Andrade
- Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
| | - Diego Menezes
- Instituto de Tecnologia e Pesquisa (ITP), Aracaju, Brazil
| | - Natália M Tavares
- Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Claudia I Brodskyn
- Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Deboraci B Prates
- Departamento de Biomorfologia, Instituto de Ciências da Saúde, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Marilda S Gonçalves
- Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Camila I de Oliveira
- Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Roque P Almeida
- Departamento de Medicina, Hospital Universitário, Universidade Federal de Sergipe (UFS), Aracaju, Brazil
| | - Marcelo T Bozza
- Departamento de Imunologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Bruno B Andrade
- Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil.,Universidade Salvador (UNIFACS), Laureate Universities, Salvador, Brazil.,Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
| | - Valeria M Borges
- Instituto Gonçalo Moniz (IGM), Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Faculdade de Medicina, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| |
Collapse
|
35
|
The Deadly Dance of B Cells with Trypanosomatids. Trends Parasitol 2017; 34:155-171. [PMID: 29089182 DOI: 10.1016/j.pt.2017.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/29/2017] [Accepted: 10/06/2017] [Indexed: 01/18/2023]
Abstract
B cells are notorious actors for the host's protection against several infectious diseases. So much so that early vaccinology seated its principles upon their long-term protective antibody secretion capabilities. Indeed, there are many examples of acute infectious diseases that are combated by functional humoral responses. However, some chronic infectious diseases actively induce immune deregulations that often lead to defective, if not deleterious, humoral immune responses. In this review we summarize how Leishmania and Trypanosoma spp. directly manipulate B cell responses to induce polyclonal B cell activation, hypergammaglobulinemia, low-specificity antibodies, limited B cell survival, and regulatory B cells, contributing therefore to immunopathology and the establishment of persistent infections.
Collapse
|
36
|
Probing the efficacy of a heterologous Leishmania/L. Viannia braziliensis recombinant enolase as a candidate vaccine to restrict the development of L. infantum in BALB/c mice. Acta Trop 2017; 171:8-16. [PMID: 28288798 DOI: 10.1016/j.actatropica.2017.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/11/2017] [Accepted: 03/09/2017] [Indexed: 11/23/2022]
Abstract
In the present study, the Leishmania braziliensis enolase protein was evaluated as a vaccine candidate against visceral leishmaniasis (VL). The DNA sequence was cloned and the recombinant protein (rEnolase) was evaluated as a vaccine, associated with saponin, as an immune adjuvant. The protective efficacy of the rEnolase plus saponin combination was investigated in BALB/c mice against Leishmania infantum infection. The results revealed that the vaccine induced higher levels of IFN-γ, IL-12, and GM-CSF when a capture ELISA and flow cytometry were performed, as well as an antileishmanial nitrite production after using in vitro stimulation with rEnolase and an antigenic Leishmania preparation. The vaccinated animals, when compared to the control groups, showed a lower parasite burden in the liver, spleen, bone marrow, and paws' draining lymph nodes when both a limiting dilution technique and RT-PCR assay were performed. In addition, these mice showed low levels of antileishmanial IL-4, IL-10, and anti-Leishmania IgG1 isotype antibodies. Partial protection was associated with IFN-γ production, which was mainly mediated by CD4+ T cells. In conclusion, the present study's data showed that the L. braziliensis enolase protein could be considered a vaccine candidate that offers heterologous protection against VL.
Collapse
|
37
|
Gardinassi LG, DeSouza-Vieira TS, da Silva NO, Garcia GR, Borges VM, Campos RNS, de Almeida RP, de Miranda Santos IKF, Saraiva EM. Molecular signatures of neutrophil extracellular traps in human visceral leishmaniasis. Parasit Vectors 2017; 10:285. [PMID: 28583201 PMCID: PMC5460406 DOI: 10.1186/s13071-017-2222-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 05/26/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Infections with parasites of the Leishmania donovani complex result in clinical outcomes that range from asymptomatic infection to severe and fatal visceral leishmaniasis (VL). Neutrophils are major players of the immune response against Leishmania, but their contribution to distinct states of infection is unknown. Gene expression data suggest the activation of the NETosis pathway during human visceral leishmaniasis. Thus, we conducted an exploratory study to evaluate NET-related molecules in retrospective sera from VL patients, asymptomatic individuals and uninfected endemic controls. RESULTS We demonstrate that VL patients and asymptomatic individuals exhibit differential regulation of molecules associated with neutrophil extracellular traps (NET). These differences were observed at the transcriptional level of genes encoding NET-associated proteins; in quantifications of cell free DNA and metalloproteinase 9; and in enzymatic activity of DNAse and elastase. Moreover, multivariate analysis resulted in class-specific signatures, and ROC curves demonstrate the ability of these molecules in discriminating asymptomatic infection from uninfected controls. CONCLUSION Molecules that are associated with NETs are differentially regulated between distinct states of infection with L. infantum, suggesting that NETs might have distinct roles depending on the clinical status of infection. Although unlikely to be exclusive for VL, these signatures can be useful to better characterize asymptomatic infections in endemic regions of this disease.
Collapse
Affiliation(s)
- Luiz Gustavo Gardinassi
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil.
| | - Thiago S DeSouza-Vieira
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Naila O da Silva
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gustavo R Garcia
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Valéria M Borges
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, BA, Brazil
| | - Roseane N S Campos
- Departamento de Medicina, Hospital Universitário, Universidade Federal de Sergipe, Aracaju, SE, Brazil
| | - Roque P de Almeida
- Departamento de Medicina, Hospital Universitário, Universidade Federal de Sergipe, Aracaju, SE, Brazil
| | - Isabel K F de Miranda Santos
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil.
| | - Elvira M Saraiva
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|