Abstract
Background:
Autoimmune thyroid diseases are the most common types of autoimmune diseases, but their physiopathology is still relatively unexplored. Genotype-tissue expression (GTEx) is a publicly available repository containing RNAseq data, including profiles from thyroid. Approximately 14.8% of these glands were affected by focal lymphocytic thyroiditis and 6.3% were annotated as Hashimoto. We interrogated these data to improve the characterization of infiltrating cells and to identify new molecular pathways active in autoimmune thyroiditis.
Materials and Methods:
Histological GTEx images of 336 thyroid samples were classified into three categories, that is, non-infiltrated thyroid, small focal infiltrated thyroid, and extensive lymphoid infiltrated thyroid. Differentially expressed genes among these categories were identified and subjected to in silico pathway enrichment analysis accordingly. CIBERSORTx deconvolution was used to characterize infiltrating cells.
Results:
As expected, most of the transcriptional changes were dependent on tissue infiltration. Upregulated genes in tissues include—in addition to lineage-specific B and T cell genes—a broad representation of inhibitory immune checkpoint receptors expressed by B and T lymphocytes. CIBERSORTx analysis identified 22 types of infiltrating cells showed that T cells predominate 3:1 over B cells in glands with small infiltrates, only by 1.7:1 in those with large infiltrates. Follicular helper and memory CD4 T cells were significantly more abundant in glands with large infiltrates (p < 0.0001), but the most prominent finding in these glands was an almost sixfold increase in the number of naive B cells (p < 0.0001). A predominance of M2 macrophages over M1 and M0 macrophages was observed in the three gland categories (p < 0.001).
Conclusions:
Analysis of transcriptomic RNA-seq profiles constitutes a rich source of information for the analysis of autoimmune tissues. High-resolution transcriptomic data analysis of thyroid glands indicates the following: (a) in all infiltrated glands, active autoimmune response coexists with suppressor counteracting mechanisms involving several inhibitory checkpoint receptor pairs, (b) glands with small infiltrates contain an unexpected relatively high proportion of B lymphocytes, and (c) in highly infiltrated glands, there is a distinct transcriptomic signature of active tertiary lymphoid organs. These results support the concept that the autoimmune response is amplified in the thyroid tissue.
Keywords: autoimmunity, immune checkpoints, thyroiditis, tolerance
Introduction
Autoimmune thyroid diseases (AITDs) are the most common organ-specific autoimmune diseases, with prevalence ranging between 2% and 5% of the population (1,2). In addition to well-established forms of thyroid autoimmunity, focal thyroiditis and subclinical thyroiditis affect up to 15–20% of the population older than 60 years (3,4). Despite extensive analysis of the immune response to thyroid autoantigens (5), the fundamental aspects of the etiology and physiopathology of AITD remain unexplained, including whether intrinsic alterations in thyroid follicular cells (TFCs) play a role in disrupting tolerance (6), and which factors determine the transition from latent focal thyroiditis to fully developed AITDs.
In the past 20 years, several new levels of regulation of the immune response have been elucidated, one of them being immune checkpoint receptors (7–10). Comparison of the transcriptomic profiles of Graves' disease (GD) and control thyroid glands led us to propose the coexistence of mechanisms responsible for activating and suppressing immune responses in autoimmune glands (11). Therefore, it was not surprising to find a partially active programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) pathway in GD, suggesting that TFCs play an active role in suppressing thyroid autoimmunity once it has been established (12). However, PD-1/PD-L1 is only one of the several regulatory checkpoints that may contribute to the maintenance of peripheral tolerance, which is why a more comprehensive investigation is required.
Genotype-tissue expression (GTEx) is an extraordinary resource in which RNA-seq data from multiple human tissues derived from over 500 cadaveric organ donors have been made available (13), including >300 thyroid gland samples. Given the high prevalence of autoimmune thyroiditis in the general population (3,4), 70 glands with histological thyroiditis were identified in this data repository. This constituted an opportunity to explore the activity of additional regulatory circuits in autoimmune thyroid glands. We interrogated GTEx data to expand the knowledge on the features of infiltrating cells, to identify new molecular pathways involved in autoimmune thyroiditis pathogenesis, and to validate our previous results in GD glands (11). In addition, the analysis of the composition of leukocyte infiltration using CIBERSORTx (14,15) provides a better understanding of the similarities and differences between incipient and well-established lymphocytic thyroiditis.
Materials and Methods
Samples, histological classification, and demographics
The GTEx database v.6 included histological data corresponding to 336 thyroid glands obtained from cadaveric organ donors. After exclusion of 4 glands containing thyroid carcinomas, the degree of lymphocytic infiltration was assessed in 332 samples using GTEx Histology Viewer utility (Supplementary Table S1); of them, matching RNA-seq raw data from 292 thyroids were downloaded from the GTEx database (Fig. 1A). This study was approved by GTEx and Hospital Vall d'Hebron ethics committees.
Differential gene expression analysis
Differential gene expression (DGE) was analyzed using DESeq2 library (v.1.18.1) for R (16) and expressed as log2-fold change. Variance stabilization was achieved using the variance-stabilizing transformation method. The median of ratios was used to correct for different sequencing depths among the samples (17). The false discovery rate was corrected using the Benjamin-Hochberg procedure.
Pathway enrichment analysis
Biological significance of differentially expressed genes (DEGs) was analyzed using Reactome Pathway Knowledge (18), which provides a list of the most represented signaling pathways for each comparison among the sample categories based on the DGE between them.
Immune infiltration profiling
Composition of thyroid leukocyte infiltrates was inferred using CIBERSORTx deconvolution software (14,15,19). Briefly, for each histologically defined category, raw gene counts were normalized to transcripts per million (TPM) and processed using CIBERSORTx algorithm for RNA-seq data. The default LM22 file was selected as the reference file to identify 22 different leukocyte subsets, which were grouped into 6 major populations (Supplementary Table S2). Differences between groups were analyzed using the Kruskal–Wallis test for nonparametric data with Dunn's test correction using GraphPad Prism® 9.3. (San Diego, CA, USA). The correlation between the abundance of the 22 leukocyte subpopulations was analyzed using R.
Results
Thyroid infiltration is more frequent in female and older patients
A trained histopathologist examined thyroid images and classified the samples into three categories: not infiltrated thyroid (NIT), small focal infiltrated thyroid (SFIT), and extensive lymphoid infiltrated thyroid (ELIT). Lymphocytic infiltration was not observed in 262 samples (79%), while different grades of infiltration were observed in 70 samples (21%), 49 (15%) of which were classified as SFIT and 21 (6%) as ELIT, most of which contained organized lymphoid follicles with germinal centers that could be considered tertiary lymphoid tissue and be suggestive of Hashimoto thyroiditis (HT). As expected, focal thyroid infiltration was more frequent in females and in donors older than 60 years (Table 1 and Fig. 1B). As the proportion of glands with large lymphocytic infiltrates was close to the estimated prevalence of HT in the United States (20), we can consider that this sample is representative of the U.S. adult population.
Table 1.
Sample information | NIT | SFIT | ELIT | p * |
---|---|---|---|---|
Sample no., n (%) | 262 (78.9) | 49 (14.8) | 21 (6.3) | |
Sex, n (%) | < 0.01 | |||
Female (n = 122) | 89 (71.8) | 21 (16.9) | 14 (11.3) | |
Male (n = 212) | 173 (83.2) | 28 (13.5) | 7 (3.4) | |
Age group, n (%) | < 0.01 | |||
<40 (n = 41) | 32 (78) | 7 (17) | 2 (5) | |
40–60 (n = 179) | 147 (82) | 10 (12) | 4 (7) | |
>60 (n = 112) | 83 (74) | 21 (19) | 8 (7) |
p Values calculated using chi-square test.
ELIT, extensive lymphoid infiltrated thyroid; GTEx, genome-tissue expression; NIT, not infiltrated thyroid; SFIT, small focal infiltrated thyroid.
DGE is highly dependent on tissue infiltration
RNA-seq data were first analyzed by principal component analysis to compare the infiltration categories. Samples corresponding to the NIT and ELIT categories were mapped to the opposite sides of the plot. SFIT samples were distributed across and overlapped with the ELIT and NIT area clusters (Fig. 2A), suggesting that the SFIT category might be a transitional state between the NIT and ELIT categories.
As expected, the transcriptomic profiles of the ELIT glands were highly enriched in lymphocyte-specific transcripts with a moderate reduction in TFCs' restricted genes. This reduction was more pronounced for genes encoding proteins that are involved in thyroid-specific functions (TG, TPO, DIO1, SLC5A5, and DUOX1) than for genes maintaining the thyroid epithelial cell structure (KRT7, KRT18, CLDN3, OCLN, and GJA19) (Table 2). All these changes were very evident in the comparison of the ELIT glands with the NIT glands, but much less evident when comparing SFIT to NIT (Fig. 2B, C and Table 3). Besides the reduction in genes specific to thyroid epithelial cells, there was an even more marked reduction in levels of calcitonin, gastrin-releasing peptide , somatostatin, and chromogranin as if C cells were more negatively affected by the lymphocytic infiltration than thyroid epithelial cells; however, there is no evidence of calcitonin deficiency in patients with thyroid autoimmune disease (21).
Table 2.
Gene | Protein coded | Tissue or cell expression | Log2 fold change | p-value adjusted |
---|---|---|---|---|
Upregulated | ||||
SERPINA9 | Serpin family A member 9 | Germinal center associated B cells | 8.838 | 4.99E-11 |
TCL1A | TCL1 family AKT co-activator A | B cells and | 7.880 | 2.47E-21 |
FCAMR | Fc fragment of IgA and IgM receptor | Lymphoid tissue, renal epithelium | 7.769 | 8.64E-14 |
FCRL4 | Fc receptor-like 4 | Epithelial memory B cells | 7.301 | 2.36E-16 |
CD19 | CD19 molecule | B cells | 7.243 | 1.43E-49 |
PAX5 | Paired box 5 | B cells | 7.240 | 1.61E-28 |
FCRL2 | Fc receptor-like 2 | B cells | 7.119 | 5.25E-27 |
FCRL1 | Fc receptor-like 1 | B cells | 7.101 | 2.08E-29 |
CR2 | Complement C3d receptor 2 | B cells | 7.038 | 9.69E-45 |
FDCSP | Follicular dendritic cell-secreted protein | Dendritic cells | 7.028 | 2.08E-07 |
Downregulated | ||||
CALCA | Calcitonin-related polypeptide alpha | Thyroid C cells | −6496 | 1.09E-04 |
GRP | Gastrin-releasing peptide | Ubiquitously expressed | −6417 | 6.39E-01 |
CHGA | Chromogranin A | Parathyroid and adrenal gland, other neuroendocrine cells | −4337 | 6.15E-03 |
SST | Somatostatin | Digestive and nervous systems, adrenal glands | −3771 | 9.72E+00 |
SCGN | Secretagogin, EF-hand calcium binding protein | Digestive and nervous systems, hypophysis | −3527 | 2.88E-04 |
VGF | VGF nerve growth factor inducible | Nervous system, hypophysis | −3418 | 8.49E-01 |
VSTM2A | V-set and transmembrane domain containing 2A |
Nervous system | −3274 | 2.32E+00 |
LOC100128317 | Uncharacterized LOC100128317 | — | −2889 | 1.77E-03 |
PTPRN | Protein tyrosine phosphatase receptor type N | Nervous system, hypophysis, adrenal glands | −2844 | 2.57E+00 |
CSMD3 | CUB and Sushi multiple domains 3 | Nervous system, hypophysis, testis | −2827 | 6.59E-03 |
Table 3.
Gene | Protein coded | Tissue or cell expression | Log2 fold change | p-Value adjusted |
---|---|---|---|---|
Upregulated | ||||
IGLL5 | Immunoglobulin lambda-like polypeptide 5 | B cells | 4.154 | 4.33E-10 |
CR2 | Complement C3d receptor 2 (CD21) | B cells | 3.903 | 2.88E-34 |
FCAMR | Fc fragment of IgA and IgM receptor | Lymphoid tissue. renal epithelium | 3.848 | 1.04E-06 |
CXCL13 | C-X-C motif chemokine ligand 13 | Secondary lymphoid organs | 3.773 | 2.30E-11 |
FCRL4 | Fc receptor-like 4 | Epithelial memory B cells | 3.767 | 1.90E-08 |
CD79A | CD79a molecule | B cells | 3.707 | 2.20E-21 |
LINC02422 | Long intergenic nonprotein coding RNA 2422 | — | 3.654 | 8.76E-08 |
SERPINA9 | Serpin family A member 9 | Germinal center associated B cells | 3.648 | 9.01E-01 |
TNFRSF13C | TNF receptor superfamily member 13C (BAFF-R) | B cells | 3.555 | 6.81E-22 |
FCRLA | Fc receptor-like A | B cells | 3.546 | 3.65E-12 |
Downregulated | ||||
THEG | Testicular haploid expressed gene protein | Spermatocytes | −2.043 | 3.88E-03 |
LINC00051 | Long intergenic nonprotein coding RNA 51 | — | −1.797 | 3.87E-03 |
LESD1 | Proteoglycan 3. proeosinophil major basic protein 2 pseudogene | Peripheral blood | −1.071 | 6.61E-03 |
USP6 | Ubiquitin specific peptidase 6 | Ubiquitously expressed | −0.989 | 7.40E-04 |
LINC02232 | Long intergenic non-protein coding RNA 2232 | — | −0.812 | 9.82E-03 |
ART5 | ADP-ribosyltransferase 5 | Testis | −0.742 | 3.10E-04 |
HSD11B2 | Hydroxysteroid 11-beta dehydrogenase 2 | Kidney, digestive system, other tissues | −0.576 | 5.77E-03 |
RGS17 | Regulator of G protein signaling 17 | Ubiquitously expressed | −0.441 | 2.68E-03 |
MDH1B | Malate dehydrogenase 1B | Testis | −0.395 | 9.07E-03 |
KLHL10 | Kelch-like family member 10 | Testis | −0.376 | 3.61E-03 |
The most DEGs specific to immune cells in SFIT and ELIT glands as opposed to those in NIT glands were mostly B lymphocyte-coding genes. Some genes were highly lineage specific (e.g., CD19, CD79A, BLK, and BTK) and other genes were highly expressed in B cells, but shared with other cell types (CR2 or BAFFR). Although not among the top DEGs, there were many upregulated T cell-specific genes in the infiltrated samples compared to those in the NIT group, many of which exhibited high-fold change values in most ELIT glands.
In silico characterization of thyroid immune infiltration
For each subpopulation identified using CIBERSORTx deconvolution software (Supplementary Table S2), its relative fraction in relation to total leukocyte infiltration as well as an absolute score that measures the overall abundance within each sample were calculated. Unexpectedly, the relative presence of each major population in the three gland categories showed that glands histologically classified as NIT contained a small, but not negligible number of leukocyte transcripts, which did not reflect the distribution of leukocyte populations in the blood, but was similar to those infiltrating the SFIT and ELIT glands. This suggests that leukocytes present in NIT glands were not cells trapped in the capillaries, but a mix of myeloid and lymphoid cells that are probably normal constituents of the thyroid tissue (Fig. 3A).
In all categories, lymphocytes were the main infiltrating population. While the proportion of T cells in the total infiltrating cells remained around 50% in the three categories, B cells showed a pronounced increase in SFIT and ELIT compared to those in NIT samples (15.0% ± 9.4% vs. 6.9% ± 4.5%, p < 0.0001 and 31.2% ± 13.0% vs. 6.9% ± 4.5%, p < 0.0001, respectively). By contrast, SFIT and ELIT samples showed a relative decrease in NK cells (6.4% ± 3.6% vs. 9.5% ± 4.4%, p < 0.0001 and 4.1% ± 3.3% vs. 9.5% ± 4.4%, p < 0.0001, respectively) and in monocytes and macrophages (26.8% ± 7.9% vs. 30.5% ± 8.3%, p < 0.05 and 16.9% ± 5.3% vs. 30.5% ± 8.3%, p < 0.0001, respectively; Fig. 3B).
To determine whether NIT, SFIT, and ELIT were quantitatively or qualitatively different, the correlations between the proportions of infiltrating leukocyte subpopulations were analyzed separately for each group of samples (Fig. 3C). In NIT samples, the relative infiltration of different leukocyte subpopulations was only weakly correlated with each other; however, multiple mutual correlations were observed in the ELIT category glands. In this category, a remarkable negative correlation was observed between the relative presence of M2 macrophages and memory B cells (r = −0.72, p < 0.01) and follicular CD4 T cells (r = −0.67, p < 0.01). A negative correlation between M1 macrophages and naive B cells (r = −0.58, p < 0.05) and a positive correlation between M1 macrophages and activated NK cells (r = 0.57, p < 0.05) were also observed (Fig. 3D). From the above, it can be deduced that the differences in the infiltrates in NIT, SFIT, and ELIT are both quantitative and qualitative.
The absolute infiltration score obtained for thyroid samples showed a significant increase of leukocytes in glands of SFIT and ELIT categories compared to that in NIT glands (1.25 ± 0.56 vs. 0.91 ± 0.20, p < 0.0001 and 2.57 ± 0.92 vs. 0.91 ± 0.20, p < 0.0001, respectively), confirming the classification generated by histopathological examination of the GTEx images (Supplementary Fig. 1A, B). Naive B cells, memory B cells, and plasma cells were significantly increased in infiltrated tissues (SFIT and ELIT) compared to those in NIT samples. While the relative proportion of total T lymphocytes showed no difference among glands of different infiltration categories, there was an increase in the absolute number of infiltrating resting CD4 memory cells, follicular helper CD4 cells, and CD8 cells in SFIT and ELIT groups compared with NIT glands. Interestingly, infiltrated glands also showed higher activation of NK cells and increased numbers of M1 and M2 macrophages, but not M0 macrophages. All these changes were especially prominent in most infiltrated glands belonging to the ELIT category (Fig. 4A and Supplementary Table S3).
Th1-associated genes are increased in infiltrated thyroid tissues
Resting CD4 memory cells were the major subpopulation present in the infiltrates of NIT, SFIT, and ELIT glands. This version of CIBERSORTx gives no information on T cell polarization, such as Th1, Th2, and Th17. To overcome this shortcoming, we investigated the DEGs associated with Th polarization in the GTEx data (22). This analysis indicated that Th1 was the predominant CD4 response associated with lymphocytic thyroiditis, which increased further in the more infiltrated tissues belonging to the ELIT category (Fig. 4B).
Immune regulatory pathways are upregulated in SFIT and ELIT samples
Six of the top 10 pathways were common between the SFIT and ELIT gland categories. These pathways included immunoregulatory interactions, co-stimulation by CD28 family molecules, cellular migration, and B cell activation (Fig. 5A and Supplementary Tables S4 and S5). Since PD-1 signaling was found to be within the 10 most enriched biological pathways in the ELIT category, but not in the SFIT category, we investigated whether the most infiltrated glands would also show increased expression of other inhibitory immune checkpoint receptors.
SFIT samples overexpressed only three inhibitory immune checkpoint coding genes with an absolute fold change greater than two: FCRL3 and FCRL4, which were expressed by B cells and TIGIT, a negative receptor mainly expressed by T cells (Fig. 5B). ELIT samples showed increased expression of a broader variety of immune checkpoint receptor genes, including PD-1, LAG3, CTLA-4, and TIM3, which are inhibitory checkpoint receptors of T lymphocytes. Among genes coding for immune checkpoints, only CD112, CD113 and CD115 showed moderate downregulation in ELIT glands when compared to that in the NIT category (Supplementary Table S6).
B cells and germinal center activity revealed
As the formation of tertiary lymphoid organs (TLOs) has been postulated to play a pathogenic role in autoimmune diseases, including thyroid autoimmune diseases (23), we specifically examined the DEGs involved in the formation and activation of lymphoid follicles. Lymphotoxin-alpha, CXCR4, CXCR5, CXCL13, and CCL19 participate in the formation of lymphoid follicles and were highly upregulated in ELIT glands, but only CXCR5 and CXCL13 were significantly upregulated in SFIT glands. The enzyme AICDA, which is essential for B cell somatic hypermutation and class switch recombination processes specific for activated germinal centers, was markedly increased in ELIT (fivefold) and SFIT glands, although by a lesser extent (1.7-fold) (Fig. 5C). In addition, SERPIN9 and CR2, genes typically expressed by centroblasts, were among the top 10 upregulated genes in SFIT and ELIT samples, as was follicular dendritic cell secreted protein (FDCSP), a gene that is specifically expressed by follicular dendritic cells.
CIBERSORTx analysis revealed a marked expansion of Tfh and memory B cells in ELIT glands, but only a very limited expansion in the SFIT category (Fig. 4B). Notably, FCRL4, which codes for an inhibitory receptor originally described in activated B cells from mucosal lymphoid tissues, is one of the DEGs in SFIT and ELIT categories.
Discussion
We report an in-depth analysis focusing on the changes associated with focal and extensive lymphocytic thyroiditis aided by public RNA-seq repositories such as GTEx. A previous analysis of the same dataset by Cho et al. (24) described transcriptomic changes in thyroid glands related to aging, including the upregulation of immune system genes. However, this was not employed in analyzing autoimmune pathogenesis. Both studies are limited by data available at the GTEx, and therefore, clinical and laboratory details relevant to thyroid pathology could not be analyzed.
We have demonstrated that majority of the most upregulated genes in samples with small infiltrates (SFIT) were specifically related to the immune system. Although this pattern was also observed in samples with extensive lymphoid infiltrates (ELIT), this category was characterized by a higher number of DEGs. Most downregulated genes in SFIT and ELIT tissues belonged to ubiquitous signaling pathways, although the expression of functional and structural genes specific to TFCs was also reduced, especially in ELIT samples. This reduction may be an indication of parenchymal destruction.
Pathway enrichment analysis confirmed that most overrepresented biological pathways in SFIT and ELIT categories were related to immune system activation and regulation. In addition, genes coding for immune checkpoint molecules expressed on B and T lymphocytes were significantly increased, including BTLA, CTLA-4, TIGIT, FCRL3, FCRL4, and PD-1 (25–27). The expression of immune checkpoints was further enhanced in the ELIT category samples, suggesting that their upregulation may be related to the degree of tissue infiltration. Antigen presentation and interferon signatures genes, which were the focus of our previous analysis of GD by array hybridization (11), are also upregulated in the ELIT category of samples. However, due to the different methodologies in the two studies, it is difficult to make a productive comparison.
Characterization of thyroid infiltration using the CIBERSORTx algorithm led to the identification of changes presumably associated with the progression of autoimmune responses. In agreement with previous reports, thyroid infiltrates were mainly composed of T and B lymphocytes (28,29). Other changes observed include an increase of M2 macrophages and the polarization of T cells toward the Th1 response, as reported previously (11,30). In addition, ELIT samples showed a significant increase in infiltrating naive and memory B cell subpopulations and follicular helper T cells, which reflect the known process of TLO formation in thyroid autoimmunity (31–33). This is further supported by the upregulation of genes involved in germinal center formation in the more infiltrated glands, including LT-α, CXCR5, CXCL13, and CCL19 (34–37).
Interestingly, among the top 10 DEGs in SFIT and ELIT, we detected that FCRL4, a low-affinity immunoglobulin A receptor with immunoregulatory activity, expressed by a subpopulation of memory B cells initially found to be restricted to mucosa-associated lymphoid tissue (38–40). However, recent studies have identified an increase of FCRL4+ B cells in TLOs formed in patients with primary Sjögren syndrome or rheumatoid arthritis, and in peripheral mononuclear cells in GD, but not in HT patients. These data suggest a possible role of this cell subset in the pathogenesis of organ-specific autoimmune diseases (41–43).
Overall, these results provide a better understanding of transcriptomic changes associated with the initiation and progression of thyroid-specific autoimmune responses. Despite the overlap in DGE observed between SFIT and ELIT samples, CIBERSORTx analysis revealed different compositions of infiltrating leukocyte subpopulations without the bias inherent to procedures that include tissue digestion. These results suggest that immune responses in autoimmunity evolve during the course of the disease toward a more chronic state, which is characterized by a better coordinated interplay between infiltrating leukocytes, the presence of TLOs, and the upregulation of immune checkpoint molecules, indicating a shift toward exhaustion of infiltrating cells (44,45). Along with previous works, these results help to understand better the mechanisms underlying thyroid-specific immune-related adverse events observed in cancer immunotherapy after treatment with anti-immune checkpoint receptors antibodies such as Nivolumab (46,47).
Changes that lead to the transition from subclinical forms of thyroiditis to Hashimoto's thyroiditis could not be clearly identified in this study. However, the data point to the recruitment of Th1 cells and the formation of TLO as critical steps that may give rise to an expansion loop that ultimately leads to clinical autoimmunity (23), being the significance of last process further supported by the therapeutic success of anti-CD20 in a large variety of autoimmune diseases, including Graves' ophthalmopathy (48,49).
As the transcriptional data obtained by RNA-seq are dominated by the infiltration itself, it accounts for most expression changes observed and possibly masks those occurring in TFCs. Additional transcriptional studies using isolated TFCs are needed to assess the role of parenchymal cells in local inflammation, and their contribution to peripheral tolerance mechanisms regulating autoimmune responses.
Supplementary Material
Acknowledgments
We thank the GTEx Project and database of Genotypes and Phenotypes (dbGaP) for granting access to the transcriptomic and clinical information of thyroid tissue donors used for this work. The GTEx Project was supported by the Common Fund of the Office of the Director of the National Institutes of Health, and by the National Cancer Institute (NCI), the National Human Genome Research Institute (NHGRI), the National Heart, Lung, and Blood Institute (NHLBI), the National Institute on Drug Abuse (NIDA), the National Institute of Mental Health (NIMH), and the National Institute of Neurological Disorders and Stroke (NINDS). The data used for the analyses described in this article were obtained from dbGaP accession number phs000424.vN.pN. We also thank Ricardo Gonzalo and the Bioinformatics and Statistics Unit (UEB) from the Vall d'Hebron Institute of Research (VHIR) for their contribution to data analysis.
Authors' Contributions
D.Á.-S. contributed to the design of the study, analyzed the data, and wrote part of the article. A.M.-S. contributed to data analysis. A.G.-B., I.B., O.G., E.C., P.M.-L., A.P., C.Z., and C.I. contributed to the review of the article and the interpretation of results. R.P.-B. contributed to the design of the study, to the interpretation of the results, wrote part of the article, and was responsible for the approval of the final draft.
Author Disclosure Statement
No competing financial interests exist.
Funding Information
This work was supported by grants PI17/00324 from the Instituto de Salud Carlos III and was co-financed by the European Regional Development Fund (ERDF). The GTEx Project was supported by the Commond Fund of the Office of the Director of the National Institutes of Health (NIH), and by NCI, NHGRI, NHLBI, NIDA, NIMH, and NINDS (13).
Supplementary Material
References
- 1. McLeod DSA, Cooper DS. 2012. The incidence and prevalence of thyroid autoimmunity. Endocrine 42:252–265. [DOI] [PubMed] [Google Scholar]
- 2. Madariaga AG, Santos Palacios S, Guillén-Grima F, Galofré JC. 2014. The incidence and prevalence of thyroid dysfunction in Europe: a meta-analysis. J Clin Endocrinol Metab 99:923–931. [DOI] [PubMed] [Google Scholar]
- 3. Kurashima C, Hirokawa K. 1985. Focal lymphocytic infiltration in thyroids of elderly people. Histopathological and immunohistochemical studies. Surv Synth Pathol Res 4:457–466. [DOI] [PubMed] [Google Scholar]
- 4. Williams ED, Doniach I. 1962. The post-mortem incidence of focal thyroiditis. J Pathol Bacteriol 83:255–264. [DOI] [PubMed] [Google Scholar]
- 5. Fröhlich E, Wahl R. 2017. Thyroid autoimmunity: role of anti-thyroid antibodies in thyroid and extra-thyroidal diseases. Front Immunol 8:51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Bottazzo GF, Hanafusa T, Pujol-Borrell R, Feldmann M. 1983. Role of aberrant Hla-Dr expression and antigen presentation in induction of endocrine autoimmunity. Lancet 322:1115–1119. [DOI] [PubMed] [Google Scholar]
- 7. Chen L, Flies DB. 2013. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol 13:227–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Nüssing S, Trapani JA, Parish IA. 2020. Revisiting T cell tolerance as a checkpoint target for cancer immunotherapy. Front Immunol 11:1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. 2002. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A 99:12293–12297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, Rosenberg SA. 2009. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 114:1537–1544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Ruiz-Riol M, Armengol Barnils MDP, Colobran Oriol R, Sánchez Pla A, Borràs Serres FE, Lucas-Martin A, Martínez Cáceres EM, Pujol-Borrell R. 2011. Analysis of the cumulative changes in Graves' disease thyroid glands points to IFN signature, plasmacytoid DCs and alternatively activated macrophages as chronicity determining factors. J Autoimmun 36:189–200. [DOI] [PubMed] [Google Scholar]
- 12. Álvarez-Sierra D, Marín-Sánchez A, Ruiz-Blázquez P, de Jesús-Gil C, Iglesias-Felip C, Gonzalez O, Casteras A, Ferrer-Costa R, Nuciforo P, Colobran R, Pujol-Borrell R. 2019. Analysis of the PD-1/PD-L1 axis in human autoimmune thyroid disease: insights into pathogenesis and clues to immunotherapy associated thyroid autoimmunity. J Autoimmun 103:102285. [DOI] [PubMed] [Google Scholar]
- 13. GTEx Consortium 2013. The Genotype-Tissue Expression (GTEx) project. Nat Genet 45:580–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu, Y, Hoang CD, Diehn M, Alizadeh AA. 2015. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods 12:453–457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Newman AM, Steen CB, Liu CL, Gentles AJ, Chaudhuri AA, Scherer F, Khodadoust MS, Esfahni MS, Luca BA, Steiner D, Diehn M, Alizadeh AA. 2019. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat Biotechnol 37:773–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Love MI, Huber W, Anders S. 2014. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 15:1–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Anders S, Huber W. 2010. Differential expression analysis for sequence count data. Genome Biol 11:R106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Fabregat A, Sidiropoulos K, Garapati P, Gillespie M, Hausmann K, Haw R, Jassal B, Jupe S, Korninger F, McKay S, Matthews L, May B, Milacic M, Rothfels K, Shamovsky V, Webber M, Weiser J, Williams M, Wu G, Stein L, Hermjakob H, D'Eustachio P. 2016. The reactome pathway knowledgebase. Nucleic Acids Res 44:D481–D487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Chen B, Khodadoust MS, Liu CL, Newman AM, Alizadeh AA. 2018. Profiling tumor infiltrating immune cells with CIBERSORT. Methods Mol Biol 1711:243–259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Staii A, Mirocha S, Todorova-Koteva K, Glinberg S, Jaume JC. 2010. Hashimoto thyroiditis is more frequent than expected when diagnosed by cytology which uncovers a pre-clinical state. Thyroid Res 3:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Grani G, Nesca A, del Sordo M, Calvanese A, Carbotta G, Bianchini M, Fumarola A. 2012. Interpretation of serum calcitonin in patients with chronic autoimmune thyroiditis. Endocr Relat Cancer 19:345–349. [DOI] [PubMed] [Google Scholar]
- 22. Radens CM, Blake D, Jewell P, Barash Y, Lynch KW. 2020. Meta-analysis of transcriptomic variation in T-cell populations reveals both variable and consistent signatures of gene expression and splicing. RNA 26:1320–1333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Aloisi F, Pujol-Borrell R. 2006. Lymphoid neogenesis in chronic inflammatory diseases. Nat Rev Immunol 6:205–217. [DOI] [PubMed] [Google Scholar]
- 24. Cho BA, Yoo SK, Song YS, Kim SJ, Lee KE, Shong M, Park YJ, Seo JS. 2018. Transcriptome network analysis reveals aging-related mitochondrial and proteasomal dysfunction and immune activation in human thyroid. Thyroid 28:656–666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Wherry EJ 2011. T cell exhaustion. Nat Immunol 12:492–499. [DOI] [PubMed] [Google Scholar]
- 26. Murphy KM, Nelson CA, Šedý JR. 2006. Balancing co-stimulation and inhibition with BTLA and HVEM. Nat Rev Immunol 6:671–681. [DOI] [PubMed] [Google Scholar]
- 27. Tz G, Ehrhardt RA, Davis RS, Hsu JT, Leu CM, Ehrhardt A, Cooper MD. 2003. The inhibitory potential of Fc receptor homolog 4 on memory B cells. Proc Natl Acad Sci U S A 100:13489–13494 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Pearce EN, Farwell AP, Braverman LE. 2003. Thyroiditis. N Engl J Med 348:2646–2655. [DOI] [PubMed] [Google Scholar]
- 29. Margolick JB, Hsu SM, Volkman DJ, Burman KD, Fauci AS. 1984. Immunohistochemical characterization of intrathyroid lymphocytes in Graves' disease. Interstitial and intraepithelial populations. Am J Med 76:815–821. [DOI] [PubMed] [Google Scholar]
- 30. Nanba T, Watanabe M, Inoue N, Iwatani Y. 2009. Increases of the Th1/Th2 cell ratio in severe Hashimoto's disease and in the proportion of Th17 cells in intractable Graves' disease. Thyroid 19:495–501. [DOI] [PubMed] [Google Scholar]
- 31. Weetman AP 2004. Cellular immune responses in autoimmune thyroid disease. Clin Endocrinology 61:405–413. [DOI] [PubMed] [Google Scholar]
- 32. Armengol MP, Juan M, Lucas-Martín A, Fernández-Figueras MT, Jaraquemada D, Gallart T, Pujol-Borrell R. 2001. Thyroid autoimmune disease: demonstration of thyroid antigen-specific B cells and recombination-activating gene expression in chemokine-containing active intrathyroidal germinal centers. Am J Pathol 159:861–873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Mohr A, Trésallet C, Monot N, Bauvois A, Abiven D, Atil M, Claër L, Malhotra R, Mayer G, Balderas R, Vaarala O, Deniziaut G, Brocheriou I, Buffet C, Leenhardt L, Gorochov G, Miyara M. 2020. Tissue infiltrating LTi—like Group 3 innate lymphoid cells and T follicular helper cells in Graves' and Hashimoto's thyroiditis. Front Immunol 11:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Tang H, Zhu M, Qiao J, Fu YX. 2017. Lymphotoxin signalling in tertiary lymphoid structures and immunotherapy. Cell Mol Immunol 14:809–818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Neyt K, Perros F, GeurtsvanKessel CH, Hammad H, Lambrecht BN. 2012. Tertiary lymphoid organs in infection and autoimmunity. Trends Immunol 33:297–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Furtado GC, Marinkovic T, Martin AP, Garin A, Hoch B, Hubner W, Chen BK, Genden E, Skobe M, Lira SA. 2007. Lymphotoxin β receptor signaling is required for inflammatory lymphangiogenesis in the thyroid. Proc Natl Acad Sci U S A 104:5026–5031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Wu B xuan, Zhao L dan, Zhang × 2019. CXCR4 and CXCR5 orchestrate dynamic germinal center reactions and may contribute to the pathogenesis of systemic lupus erythematosus. Cell Mol Immunol 16:724–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Davis RS, Wang Y, Kubagawa H, Cooper MD. 2001. Identification of a family of Fc receptor homologs with preferential B cell expression. Proc Natl Acad Sci U S A 98:9772–9777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Liu Y, Goroshko S, Leung LYT, Dong S, Khan S, Campisi P, Propst EK, Wolter NE, Grunebaum E, Ehrhardt GRA. 2020. FCRL4 Is an Fc receptor for systemic IgA, but not mucosal secretory IgA. J Immunol 205:533–538. [DOI] [PubMed] [Google Scholar]
- 40. Falini B, Tiacci E, Pucciarini A, Bigerna B, Kurth J, Hatzivassiliou G, Droetto S, Verducci-Galleti B, Gambacorta M, Orazi A, Pasqualucci L, Miller I, Kuppers R, Dalla-Favera R, Cattoretti G. 2003. Expression of the IRTA1 receptor identifies intraepithelial and subepithelial marginal zone B cells of the mucosa-associated lymphoid tissue (MALT). Blood 102:3684–3692. [DOI] [PubMed] [Google Scholar]
- 41. Verstappen GM, Ice JA, Bootsma H, Pringle S, Haacke EA, de Lange K, van der Vries GE, Hickey P, Vissink A, Spijkervet FKL, Lessard CJ, Kroese FGM. 2020. Gene expression profiling of epithelium-associated FcRL4+ B cells in primary Sjögren's syndrome reveals a pathogenic signature. J Autoimmun 109:102439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Spengler J, Amara K, Clay E, Yeo L, Ramsköld D, Spengler J, Sippl N, Cameron JA, Israelsson, Titcombe PJ, Grönwall C, Sahbudin I, Filer A, Raza K, Malmström V, Scheel-Toellner D. 2017. B cells expressing the IgA receptor FcRL4 participate in the autoimmune response in patients with rheumatoid arthritis. J Autoimmun 81:34–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Rostamzadeh D, Dabbaghmanesh MH, Shabani M, Hosseini A, Amirghofran Z. 2015. Expression profile of human Fc receptor-like 1, 2, and 4 molecules in peripheral blood mononuclear cells of patients with Hashimoto's thyroiditis and Graves' disease. Horm Metab Res 47:693–698. [DOI] [PubMed] [Google Scholar]
- 44. McKinney EF, Smith KGC 2016 T cell exhaustion: understanding the interface of chronic viral and autoinflammatory diseases. Immunol Cell Biol 1–22. [DOI] [PubMed] [Google Scholar]
- 45. Sharpe AH, Pauken KE. 2018. The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol 18:153–167. [DOI] [PubMed] [Google Scholar]
- 46. Byun DJ, Wolchok JD, Rosenberg LM, Girotra M. 2017. Cancer immunotherapy—immune checkpoint blockade and associated endocrinopathies. Nat Rev Endocrinol 13:195–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Chang LS, Barroso-Sousa R, Tolaney SM, Hodi FS, Kaiser UB, Min L. 2018. Endocrine toxicity of cancer immunotherapy targeting immune checkpoints. Endocr Rev 40:17–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. GRAID investigators 2011. Safety and clinical outcomes of rituximab therapy in patients with different autoimmune diseases: experience from a national registry (GRAID). Arthritis Res Ther 13:R75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Chen J, Chen G, Sun H. 2000. Intravenous rituximab therapy for active Graves' ophthalmopathy: a meta-analysis. Hormones 20:279–286. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.