Cargando…

Identification of Critical Transcriptomic Signaling Pathways in Patients with H Syndrome and Rosai-Dorfman Disease

Biallelic mutations in SLC29A3 cause histiocytosis-lymphadenopathy plus syndrome, also known as H syndrome (HS). HS is a complex disorder, with ~ 25% of patients developing autoinflammatory complications consisting of unexplained fevers, persistently elevated inflammatory markers, and unusual lympha...

Descripción completa

Detalles Bibliográficos
Autores principales: Lara-Reyna, Samuel, Poulter, James A., Vasconcelos, Elton J.R., Kacar, Mark, McDermott, Michael F., Tooze, Reuben, Doffinger, Rainer, Savic, Sinisa
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Springer US 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7858559/
https://www.ncbi.nlm.nih.gov/pubmed/33284430
http://dx.doi.org/10.1007/s10875-020-00932-1
_version_ 1783646624944226304
author Lara-Reyna, Samuel
Poulter, James A.
Vasconcelos, Elton J.R.
Kacar, Mark
McDermott, Michael F.
Tooze, Reuben
Doffinger, Rainer
Savic, Sinisa
author_facet Lara-Reyna, Samuel
Poulter, James A.
Vasconcelos, Elton J.R.
Kacar, Mark
McDermott, Michael F.
Tooze, Reuben
Doffinger, Rainer
Savic, Sinisa
author_sort Lara-Reyna, Samuel
collection PubMed
description Biallelic mutations in SLC29A3 cause histiocytosis-lymphadenopathy plus syndrome, also known as H syndrome (HS). HS is a complex disorder, with ~ 25% of patients developing autoinflammatory complications consisting of unexplained fevers, persistently elevated inflammatory markers, and unusual lymphadenopathies, with infiltrating CD68(+), S100(+), and CD1a(−) histiocytes, resembling the immunophenotype found in Rosai-Dorfman disease (RDD). We investigated the transcriptomic profiles of monocytes, non-activated (M0), classically activated (M1), and alternatively activated macrophages (M2) in two patients with HS, one without autoinflammatory (HS1) and one with autoinflammatory complications (HS2). RNA sequencing revealed a dysregulated transcriptomic profile in both HS patients compared to healthy controls (HC). HS2, when compared to HS1, had several differentially expressed genes, including genes associated with lymphocytic-histiocytic predominance (e.g. NINL) and chronic immune activation (e.g. B2M). The transcriptomic and cytokine profiles of HS patients were comparable to patients with SAID with high levels of TNF. SERPINA1 gene expression was found to be upregulated in all patients studied. Moreover, higher levels of IFNγ were found in the serum of both HS patients when compared to HC. Gene ontology (GO) enrichment analysis of the DEGs in HS patients revealed the terms “type I IFN,” “IFNγ signaling pathway,” and “immune responses” as the top 3 most significant terms for monocytes. Gene expression analysis of lymph node biopsies from sporadic and H syndrome-associated RDD suggests common underlying pathological process. In conclusion, monocytes and macrophages from both HS patients showed transcriptomic profiles similar to SAIDs and also uniquely upregulated IFNγ signature. These findings may help find better therapeutic options for this rare disorder. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s10875-020-00932-1.
format Online
Article
Text
id pubmed-7858559
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Springer US
record_format MEDLINE/PubMed
spelling pubmed-78585592021-02-11 Identification of Critical Transcriptomic Signaling Pathways in Patients with H Syndrome and Rosai-Dorfman Disease Lara-Reyna, Samuel Poulter, James A. Vasconcelos, Elton J.R. Kacar, Mark McDermott, Michael F. Tooze, Reuben Doffinger, Rainer Savic, Sinisa J Clin Immunol Original Article Biallelic mutations in SLC29A3 cause histiocytosis-lymphadenopathy plus syndrome, also known as H syndrome (HS). HS is a complex disorder, with ~ 25% of patients developing autoinflammatory complications consisting of unexplained fevers, persistently elevated inflammatory markers, and unusual lymphadenopathies, with infiltrating CD68(+), S100(+), and CD1a(−) histiocytes, resembling the immunophenotype found in Rosai-Dorfman disease (RDD). We investigated the transcriptomic profiles of monocytes, non-activated (M0), classically activated (M1), and alternatively activated macrophages (M2) in two patients with HS, one without autoinflammatory (HS1) and one with autoinflammatory complications (HS2). RNA sequencing revealed a dysregulated transcriptomic profile in both HS patients compared to healthy controls (HC). HS2, when compared to HS1, had several differentially expressed genes, including genes associated with lymphocytic-histiocytic predominance (e.g. NINL) and chronic immune activation (e.g. B2M). The transcriptomic and cytokine profiles of HS patients were comparable to patients with SAID with high levels of TNF. SERPINA1 gene expression was found to be upregulated in all patients studied. Moreover, higher levels of IFNγ were found in the serum of both HS patients when compared to HC. Gene ontology (GO) enrichment analysis of the DEGs in HS patients revealed the terms “type I IFN,” “IFNγ signaling pathway,” and “immune responses” as the top 3 most significant terms for monocytes. Gene expression analysis of lymph node biopsies from sporadic and H syndrome-associated RDD suggests common underlying pathological process. In conclusion, monocytes and macrophages from both HS patients showed transcriptomic profiles similar to SAIDs and also uniquely upregulated IFNγ signature. These findings may help find better therapeutic options for this rare disorder. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s10875-020-00932-1. Springer US 2020-12-07 2021 /pmc/articles/PMC7858559/ /pubmed/33284430 http://dx.doi.org/10.1007/s10875-020-00932-1 Text en © The Author(s) 2020 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
spellingShingle Original Article
Lara-Reyna, Samuel
Poulter, James A.
Vasconcelos, Elton J.R.
Kacar, Mark
McDermott, Michael F.
Tooze, Reuben
Doffinger, Rainer
Savic, Sinisa
Identification of Critical Transcriptomic Signaling Pathways in Patients with H Syndrome and Rosai-Dorfman Disease
title Identification of Critical Transcriptomic Signaling Pathways in Patients with H Syndrome and Rosai-Dorfman Disease
title_full Identification of Critical Transcriptomic Signaling Pathways in Patients with H Syndrome and Rosai-Dorfman Disease
title_fullStr Identification of Critical Transcriptomic Signaling Pathways in Patients with H Syndrome and Rosai-Dorfman Disease
title_full_unstemmed Identification of Critical Transcriptomic Signaling Pathways in Patients with H Syndrome and Rosai-Dorfman Disease
title_short Identification of Critical Transcriptomic Signaling Pathways in Patients with H Syndrome and Rosai-Dorfman Disease
title_sort identification of critical transcriptomic signaling pathways in patients with h syndrome and rosai-dorfman disease
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7858559/
https://www.ncbi.nlm.nih.gov/pubmed/33284430
http://dx.doi.org/10.1007/s10875-020-00932-1
work_keys_str_mv AT larareynasamuel identificationofcriticaltranscriptomicsignalingpathwaysinpatientswithhsyndromeandrosaidorfmandisease
AT poulterjamesa identificationofcriticaltranscriptomicsignalingpathwaysinpatientswithhsyndromeandrosaidorfmandisease
AT vasconceloseltonjr identificationofcriticaltranscriptomicsignalingpathwaysinpatientswithhsyndromeandrosaidorfmandisease
AT kacarmark identificationofcriticaltranscriptomicsignalingpathwaysinpatientswithhsyndromeandrosaidorfmandisease
AT mcdermottmichaelf identificationofcriticaltranscriptomicsignalingpathwaysinpatientswithhsyndromeandrosaidorfmandisease
AT toozereuben identificationofcriticaltranscriptomicsignalingpathwaysinpatientswithhsyndromeandrosaidorfmandisease
AT doffingerrainer identificationofcriticaltranscriptomicsignalingpathwaysinpatientswithhsyndromeandrosaidorfmandisease
AT savicsinisa identificationofcriticaltranscriptomicsignalingpathwaysinpatientswithhsyndromeandrosaidorfmandisease