RT Journal Article SR Electronic T1 Localized skin inflammation during cutaneous leishmaniasis drives a chronic, systemic IFN-γ signature JF medRxiv FD Cold Spring Harbor Laboratory Press SP 2020.12.03.20240978 DO 10.1101/2020.12.03.20240978 A1 Camila Farias Amorim A1 Fernanda O. Novais A1 Ba T. Nguyen A1 Mauricio T. Nascimento A1 Jamile Lago A1 Alexsandro S. Lago A1 Lucas P. Carvalho A1 Daniel P. Beiting A1 Phillip Scott YR 2020 UL http://medrxiv.org/content/early/2020/12/04/2020.12.03.20240978.abstract AB 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, a transcriptional analysis of lesions from patients identified the presence of a strong interferon stimulated gene (ISG) signature. To determine what systemic responses are occurring that might influence the disease, we performed RNA sequencing (RNA-seq) on the blood of L. braziliensis-infected patients, as well as healthy controls. Functional enrichment analysis identified a transcriptional ISG signature as the dominant response in the blood of patients. An increase in monocytes and macrophages in the blood, estimated from our RNA-seq dataset, was positively correlated with this ISG signature. Consistent with this result, patients had circulating IFN-γ in their serum. A cytotoxicity signature, which is a dominant feature in the lesions, was also found 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.Author summary Cutaneous leishmaniasis caused by the protozoan Leishmania braziliensis exhibits two dominant inflammatory responses in cutaneous lesions: Interferon-γ (IFN-γ)-mediated signaling, which promotes parasite control, and cytolysis mediated by cytotoxic CD8+ T and NK cells, which promotes increased pathology. To determine if these responses were limited to cutaneous lesions, we performed RNA-seq on the blood of cutaneous leishmaniasis (CL) patients, and detected both transcriptional signatures in the peripheral blood. The presence of interferon stimulated genes, as well as circulating IFN-γ, suggests that protective immune responses are not limited to the lesion site, but are occurring systemically. This may be one mechanism to ensure optimal control of the parasites, both by limiting their systemic spread and by pre-activating cells to kill the parasites prior to entry into the lesions. The cytolytic transcriptional signature was uniquely detectable in the blood of L. braziliensis patients when compared to the blood of patients with tuberculosis (TB) or malaria, further emphasizing the importance of this pathway in cutaneous leishmaniasis. Taken together, these data suggest that this localized infection has a systemic component that may have an impact the development of the disease.Competing Interest StatementThe authors have declared no competing interest.Funding StatementPS is funded by the National Institutes of Health (NIH) R01-AI-149456 (https://grants.nih.gov/grants/funding/r01.htm). PS and LPC are funded by the National Institute of Allergy and Infectious Diseases, Tropical Medicine Research Centers (TMRC) AI 136032 (https://www.niaid.nih.gov/research/tmrc-program-overview). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:This study was approved by the University of Pennsylvania's Institutional Review Board on December 8, 2015 (#823847) and the Federal University of Bahia's Committee on Ethics in Research on February 8, 2015 (#952.621). Informed consent for the collection of samples and subsequent analysis was obtained.All necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived.YesI understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).YesI have followed all appropriate research reporting guidelines and uploaded the relevant EQUATOR Network research reporting checklist(s) and other pertinent material as supplementary files, if applicable.YesOnce this manuscript is officially published, the CL peripheral blood raw sequence data will be available on the NCBI Gene expression Omnibus (GEO). CL lesion biopsy raw sequence data can be found on GEO accession GSE127831. https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE127831