Abstract
Studying the human immune system is challenging. These challenges stem from the complexity of the immune system itself, the heterogeneity of the immune system between individuals, and the many factors that lead to this heterogeneity including the influence of genetics, environment, and immune experience. Studies of the human immune system in the context of disease are increased in complexity as multiple combinations and variation in immune pathways can lead to a single disease. Thus, although individuals with a disease may share clinical features, the underlying disease mechanisms and resulting pathophysiology can be diverse among individuals with the same disease diagnosis. This has consequences for treatment of diseases as no single therapy will work for everyone, therapeutic efficacy varies among patients, and targeting a single immune pathway is rarely 100% effective. This review discusses how to address these challenges by identifying and managing the sources of variation, improving access to high quality well-curated biological samples by building cohorts, applying new technologies such as single cell omics and imaging technologies to interrogate samples and bringing to bear computational expertise in conjunction with immunologists and clinicians to interpret those results. The review has a focus on autoimmune disease, including rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus and type 1 diabetes, but its recommendations are also applicable to studies of other immune-mediated diseases.
Keywords: translational immunology, autoimmune disease, rheumatoid arthritis, Omics, antigen-specificity
Graphical Abstract
Studying the human immune system requires a toolkit that includes high quality, well-curated biological samples from carefully designed cohorts, and cutting-edge single cell approaches such as omics profiling, imaging techniques and antigen-specific assays. Data analysis and interpretation requires a research team combining expertise in bioinformatics, computational biology, immunology and clinical care. Created with BioRender.com.
Introduction
The goal of biomedical research is to understand biological systems so that we can determine the pathogenic mechanisms of disease and intervene in those processes. However, biology is complicated, and the immune system is no exception. To limit the complexity that is found in humans, scientists have relied heavily on model systems, in particular mice and immortalized cell lines, to tackle the fundamental questions of how the immune system works. These studies have yielded a deep understanding of how the components of the immune system function and have revealed genetic and biochemical mechanisms that underpin them. In addition, murine models of immune diseases have paved the way for the development of many immune therapies, from cytokine blocking biologics for the treatment of autoimmune and allergic diseases to immune checkpoint inhibitors for treatment of cancer. Yet, murine models fall short in providing us with the knowledge needed to understand the complex genetic, environmental, and experiential factors that influence the human immune system. The complexity of human immunology makes its study a daunting task, yet if we are to ultimately prevent and cure immune related diseases, it must be undertaken.
In this review, I will focus on why studying the human immune system is necessary, the hurdles that must be overcome to do it and how by reimagining the way we study the human immune system we can accelerate our ability to improve human health. Much of the discussion will focus on how this paradigm shift will impact the study of autoimmune diseases, but these same concepts and approaches can be applied to other immune diseases from allergy or infection to cancer.
The problem as illustrated through the lens of rheumatoid arthritis
Every person is unique. This is true in health as well as disease. Studies of healthy individuals have shown that immune variation between individuals is high, but that immune characteristics within an individual are stable [1]. It is also well-established that multiple factors influence an individual’s baseline immune landscape or “immunotype”. This has been elegantly shown in twin studies where shared immune features in monozygotic, but not dizygotic, twins demonstrate a clear role for genetics, conversely, features that are not shared in monozygotic twins indicate a role for environmental or stochastic factors in determining these specific features [2, 3]. Further study of shared immune phenotypes in dizygotic twins have underscored the importance of shared childhood environments in the development of an individual’s immune landscape [4]. Immune experience adds another layer to human immune heterogeneity as immunotype is influenced by many factors such as age, cytomegalovirus (CMV) status, and chronic inflammatory states [5]. Collectively these findings highlight the need to understand the breadth of immunotypes seen in health, the factors that drive differences across individuals, so that we can ultimately determine where individuals with autoimmunity deviate from immune health.
When studying autoimmune disease in humans, we must consider both the complexity of the pathogenic mechanisms that lead to disease and the immune heterogeneity between individuals with disease (Figure 1). The breadth of immune heterogeneity when viewed in the context of a complex disease like autoimmune disease means that there are multiple combinations and variation in immune pathways that can lead to a single disease. Thus, although individuals with a disease may share clinical features, the underlying disease mechanisms and resulting pathophysiology can be diverse among individuals with the same disease diagnosis. This is exemplified by the natural history of these diseases, the heterogeneity in the response to therapeutic interventions, and the complexity of the genetics of autoimmune diseases.
Figure 1. Genes, immune experience and stochastic factors drive immune heterogeneity.
A single clinical diagnosis can be caused by a combination of factors. The result is that individuals with the same clinical diagnosis are immunologically as well as clinically heterogeneous. Dissecting this heterogeneity will allow us to categorize individual patients and provide improved targeted therapies.
Rheumatoid arthritis (RA) is an example of an autoimmune disease with complexity and heterogeneity. RA, an inflammatory disease characterized by pain and swelling in the joints, is more common in women than men with a 3:1 ratio and can occur at any age, but increases in frequency with age [6]. Many, but not all, individuals with RA have anti-citrullinated protein antibodies (ACPA) and/or rheumatoid factor (RF) [7]. In addition, some individuals may have mild disease, whereas others have severe disease resulting in progressive disability due to joint damage [6]. Genetic factors are known to influence risk for RA. The strongest association is with HLA Class II alleles, for example, the HLA DRB1*0401 increases risk 6-fold [8] but genome-wide association studies (GWAS) have also identified more than 100 genetic variants that confer a modest level of risk [9, 10]. Many of these variants are in genes known to be important in the immune response, including PTPN22, which influences intracellular signaling in immune cells, CTLA4, an immune checkpoint inhibitor, and IL6R, TYK2, STAT4, all key components of cytokine signaling pathways regulating the immune response [11]. Interestingly, risk variants in HLA-DR4 and PTPN22 are linked to the development of ACPA, indicating a mechanism by which they promote autoimmunity in RA. However, genetic heritability only accounts for 60% of disease risk in RA [12–14], indicating a role for environmental factors as well. One environmental factor linked to RA is smoking, which can increase risk two-fold [15]. Also noteworthy is that genetic and environmental factors may interact to promote disease. For example, smoking enhances the combined effect of risk variants in HLA-DR4 and PTPN22 [16]. Lastly, the microbiome has also been implicated as a contributing factor to pathogenesis of RA, adding yet another level of disease complexity and individual heterogeneity [17–19].
Importantly, the impact of disease complexity overlaid with immune heterogeneity results in real-world problems in the treatment of patients with autoimmune disease. No single therapy works for everyone, therapeutic efficacy varies among patients, and targeting a single immune pathway is rarely 100% effective. Again, turning to RA, 30–40% of treatment naïve patients do not have adequate responses to current therapies [20–25]. This is despite tremendous advances in the treatment of RA and the growing numbers of types of drugs available in the clinic. The struggles that we face in treating RA likely derive, in part, from a multiplicity of disease endotypes, each driven by different combinations of immune pathways, thus requiring interventions that specifically target the culprit pathway(s). Moreover, due to the chronic nature of autoimmune diseases, time may also alter the pathogenic features of an individual’s disease. Due to the multiplicity of pathways involved, RA requires us to consider addressing these factors through sequential or combination therapies. Furthermore, like RA, other autoimmune diseases demonstrate similar complexity and heterogeneity with regards to pathogenic mechanisms. Multiple sclerosis (MS) and systemic lupus erythematosus (SLE) have a similar number ( >100) of genetic markers associated with disease [26–30], and multiple environmental factors have been linked to disease risk, including geographic location for MS [31] and air pollution for SLE [32], as well as the bacteria and viruses such as Epstein-Barr virus (EBV) [33, 34]. Similar to RA, there is heterogeneity in the response to immune interventions across the spectrum of autoimmune disease as exemplified in the recent trial of anifrolumab, a monoclonal antibody against type 1 interferon receptor subunit 1 in SLE, where only 47.8% of treated subjects showed a response [35]. Thus, to tackle the real-world problems of treating and ultimately curing autoimmune diseases in humans, we need to embrace the complexity and heterogeneity in order to develop approaches that will allow us to define the pathways and interactions that promote disease within and across individuals.
Overcoming the limitations posed by studying humans
How do we embrace complexity and heterogeneity in the study of human autoimmune diseases? First and foremost, we need to identify the sources of variation and then manage their impact through study design and data interpretation. The two main types of variation with the potential to influence the immune landscape in humans are related to sampling and intraindividual variation (Figure 2). Sampling-related variables have a direct impact on data quality and validity. The very act of obtaining a biological sample and exposing it to external factors is the first source of variation. Sample processing and storage can introduce further alterations and variation. The impact of these processes on potential immunologic readouts should be well understood and considered in study design. To minimize this variation within a given study, it is essential that all samples are handled in the same manner. Ideally, samples should be processed immediately by a dedicated lab with optimized and validated protocols. However, when this is not possible, sample types and assays that tolerate longer delays in processing should be selected and protocols for processing on site simplified to remove variability based on the site or the technician performing the isolation and storage. Importantly, for each study, the ideal protocol based on sample and assay type should be determined and, where possible, used across institutions and studies to allow for comparisons. Factors known to result in intraindividual variation should be considered as well. For example, time of day or year should be considered as a variable needing to be controlled, as there is growing recognition that diurnal variation and seasonality influence the immune landscape and responses within an individual [36–38]. At a minimum, these factors can be controlled for by tracking the time of day and date for each sample collected and considering them as variables during data analysis. However, in the ideal study design, the timing of collection would be consistent across individuals and if a longitudinal study, then at the same time of day for each time point from the same subject.
Figure 2. Designing broad scope versus in-depth studies of human immunology.
Considerations include study questions, subject selection, sample handling and data acquisition.
Subject specific features such as age, sex, race, genetics, and geographic location can lead to interindividual variation. Disease characteristics such as heterogeneity in clinical presentation and course are also sources of interindividual variation. Thus, these subject-specific variables must also be considered in the design and data capture of human studies. The majority of these data can be obtained through the electronic medical record. However, additional processes may need to be put in place to acquire more in-depth, point-of-care information on disease activity and health history. For these processes to be effective and sustainable, they need to be designed to minimize the burden on both the patient and the health care provider, while maximizing the value to the researchers. An overlooked set of subject-related variables that are of increasing importance and interest are socioeconomic and lifestyle related factors. There is a growing understanding that our immune system is influenced by diet, activity, stress, and sleep, factors that are also influenced by socioeconomic factors [39–43]. Again, as with all the variables mentioned above, tracking these variables and including them in the data analysis is critical for the generation of biologically-meaningful data that can be shared with the greater scientific community.
Human Immunology Toolkit for autoimmune diseases
Advances in understanding human immune diseases require improving access to high quality, well-curated biological samples by building cohorts, applying new technologies to interrogate samples, and bringing to bear computational expertise in conjunction with immunologists and clinicians to interpret those results.
Cohort development:
In the past 20 years, many biorepositories have been established and are rich resources for the study of human disease. Examples include the US Department of Defense Serum Repository, which was recently used to demonstrate that EBV infection increased the risk of developing MS [33] and the UK Biobank providing genetic and health information for numerous GWAS focused on autoimmune diseases [10, 44, 45]. Other biorepositories have focused on prospective longitudinal sampling in at-risk cohorts in order to understand the natural history of autoimmune disease, such as those focused on type 1 diabetes in at-risk children (Type 1 Diabetes TrialNet Pathway to Prevention Study [46, 47], TEDDY Study [48], and the BABYDIAB Study [49, 50]). These studies have given us the tools to predict who is high risk for T1D [51, 52], leading to the development of interventions to delay [53], and in the future to find therapies that will prevent, T1D. Prospective longitudinal at-risk cohorts have also been established for RA (SERA [54], SCREEN-RA [55]), SLE (SisSLE [56, 57]), and Crohn’s disease [58]. We need to continue to expand on this early success. As part of this we need to focus on inclusion of historically underrepresented groups. It is clear that disease characteristics, severity, and responses to therapy differ across ethnic, racial, and socioeconomic groups [59, 60]. An example of this is the high burden of MS in the US Black community that has been under recognized in the past [61]. Expanding the communities whom we study will allow us to understand more fully the breadth, immune mechanisms, and clinical features of immune diseases. We also need to prioritize tissue collection where possible, as this will allow us to develop better understanding of disease pathology at the site of inflammation. Ideally tissue collection should be paired with peripheral blood collection, so that we can determine biomarkers that can translate directly to the clinic. Studies of tissues have enriched our understanding of tissue resident cells [62], the patchy nature of islet inflammation in T1D [63–65], and the diversity of synovial inflammation in RA [66]. Development of additional longitudinal cohorts will also be vital for understanding how immunologic characteristics change over time and in the context of disease activity, immune experience, and environment. These longitudinal cohorts require a commitment to long term maintenance and follow-up.
Cohort selection and development and the approach to manage variation should be based on the study goals and the questions to be addressed (Figure 2). Large diverse populations and/or longitudinal cohorts are best for studies with broad scope addressing questions focused on identification of factors that influence disease development, biomarker discovery or understanding disease heterogeneity. Studies investigating the factors driving heterogeneity would require a cohort that reflects the breadth of individuals with a disease, and in this setting, well-matched control cohorts are vital. In contrast, fewer subjects but more selective recruitment based on features of disease, genetics and environmental factors are more appropriate for in-depth studies investigating specific groups of patients, disease mechanisms or the impact of interventions on these mechanisms. For example, the study of a single gene variant may require holding race and other genetic variants constant. Additionally, these in-depth studies provide an opportunity for more complex sampling protocols including collection of multiple sample types and larger sample volumes. With regards to choosing controls, again the study goals and questions are paramount since it has become increasingly evident that a healthy control may not always be the best option. Additionally, consideration must be given to which features should be matched between the controls and study subjects; age, sex, and race are the first choice but specific genetic variants and/or environmental factors may also be relevant. The type of cohort will also differ based on the scope of the study. A broad scope will require large diverse populations, excellent data management, and may require simplified and targeted sample acquisition across the cohort. An example of this is the NIH-sponsored All of Us Research Program with its goal of recruiting one million participants across races, ethnicities, age groups, US geographic regions, disease types, and socioeconomic status [67]. Whereas studies where the intent is to perform deep immune profiling will require well-curated, high-quality samples with linked genetic, immunophenotyping and functional analysis, and may have fewer subjects due to scope and cost. An example of this approach is the Milieu Interieur Project focused on defining the naturally-occurring variability of the human immune response in one thousand healthy individuals [68]. Specialized cohorts of individuals with rare or monogenic diseases are also useful to highlight specific immune pathways and processes. Additionally, cohorts of individuals treated with immune interventions can be a resource to understand immune perturbations effects in vivo; this has been a focus for many of the clinical trials conducted by both the Immune Tolerance Network and the Type 1 Diabetes TrialNet [69–71]. Lastly, Experimental Medicine Units (EMU) allow short mechanistic studies with in-depth immune monitoring and targeted immunologic endpoints [72–74].
Single Cell Technologies:
Recent advances in single cell technologies and computational methods for data analysis are moving human immunology from descriptive to mechanistic. Single cell profiling is providing unprecedented insight into the fundamentals of human immunology with the discovery of new cell types, characterization of cell-cell interactions, and lineage trajectories and is allowing us to characterize pathogenic cell types [75]. Notably, the information generated through single cell studies has the potential to identify rare pathogenic cell population or changes to cell populations that are predictive of development of disease and/or disease progression. This knowledge is essential for the development of therapies specifically targeting the cell populations and pathways involved in pathogenesis, as well as for the identification of biomarkers that predict disease progression and response to therapy.
Omics Based Approaches:
There has been an explosive growth in the number of available single cell technologies giving us the ability to assess the following at the single cell level: cell surface proteome, transcriptome, chromatin accessibility, and DNA methylation, as well as the metabolome and lipidome [75]. The most recent advance has been the development of multimodal assays for simultaneous measurement of two or more data types [75–85](Table 1). These approaches have already yielded important insights. Combining single cell RNA sequencing (scRNA-seq) and genotype data has enabled mapping of expression quantitative trait loci (eQTL) to specific cell types in several autoimmune diseases, including SLE [86, 87]. scRNA-seq has discovered a novel cell type in the blood and joints of RA patients at the time of flare [88] and cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) has recently been used to define signatures of disease activity in SLE [89]. A recent study used a trimodal approach to map the immune landscape in the human tonsil and then showed that autoimmune-associated genetic variants were enriched in chromatin accessibility regions specific to germinal center-associated T and B cells [90].
Table 1.
Multimodal single cell profiling assays
Multimodal assays | Assays | References |
---|---|---|
Cell surface protein epitopes + transcriptome | CITE-seq | 77 |
REAP-seq | 78 | |
Transcriptome + chromatin accessibility | sci-CAR | 79 |
SNARE-seq | 80 | |
SHARE-seq | 81 | |
Cell surface protein epitopes + intracellular proteins + chromatin accessibility + mitochondrial DNA variants | ASAP-seq | 82 |
Cell surface protein epitopes + chromatin accessibility | ICICLE-seq | 83 |
Cell surface protein epitopes + transcriptome + chromatin accessibility + mitochondrial DNA | DOGMA-seq | 82 |
TEA-seq | 83 | |
Cell surface protein epitopes + histone modifications | scCUT&Tag | 84 |
Transcriptome + histone modifications | Paired-Tag | 85 |
ASAP-seq: ATAC with select antigen profiling by sequencing; scATAC-seq: single cell assay for transposase-accessible chromatin using sequencing; CITE-seq: cellular indexing of transcriptomes and epitopes by sequencing; DOGMA-seq: done overfitting genomics methods acronym; ICICLE-seq: integrated cellular indexing of chromatin landscape and epitopes; Paired-Tag: parallel analysis of individual cells for RNA expression and DNA from targeted tagmentation by sequencing; REAP-seq: RNA expression and protein sequencing; scCUT&Tag: single cell cleavage under targets and tagmentation; sci-CAR: single-cell combinatorial indexing chromatin accessibility and mRNA; scRNA-seq: single cell RNA sequencing; SHARE-seq: simultaneous high-throughput ATAC and RNA expression with sequencing; SNARE-seq: single-nucleus chromatin accessibility and mRNA expression sequencing; TEA-seq: simultaneously measures transcriptomics (scRNA-seq), epitopes and chromatin accessibility (scATAC-seq)
Antigen-Specific Cell Assays:
The development of assays to identify and characterize antigen-specific T and B cells at the single cell level has also moved the field of human immunology forward. For T cells these approaches include MHC multimers [91–93] and the development of HLA agnostic approaches, such as the activation-induced marker assays [94, 95]. Similar approaches are being used to identify antigen-specific B cells [96, 97]. Importantly, these assays are being combined with single cell profiling techniques enabling an in-depth characterization of antigen-specific cells. This now allows us to identify specificity, clonal expansion, and fate mapping of antigen-specific cells [94, 98–102].
Imaging Techniques:
Imaging techniques are also important tools for studying human immunology. Whole body imaging such as magnetic resonance imaging (MRI), single photon computerized tomography (SPECT), positron emission tomography (PET) and magnetic particle imaging (MPI) are being deployed to track immune cells in vivo [103–105]. Each of these modalities rely on labelling the immune cell of interest with a tracer for visualization. To date, most of this work in humans has focused on molecular imaging of cytotoxic T cells using lineage tracers to assess the response to CAR T cell therapy or immune checkpoint inhibitor therapy in the setting of cancer [103, 105]. Whole body imaging has also been utilized in the setting of autoimmune disease but primarily in animal models. For example, SPECT imaging with nanobodies has been used to monitor macrophages in joint inflammation in the collagen-induced arthritis mouse model of RA [106, 107]. Exciting advances are also being made in the field of tissue imaging with new approaches such as spatial transcriptomics providing an opportunity to map the tissue at a depth not possible with traditional histology and immunohistochemistry. Spatial transcriptomics is unbiased as it profiles the whole transcriptome unlike immunohistochemistry or in situ hybridization, which both require a known target. Relevant to the autoimmunity field, spatial transcriptomics has provided new insight into the cell populations present in synovial tissue in RA during early disease, disease flares and remission [108–110]. It has also been utilized on postmortem brain tissue to track neurodegeneration in progressive MS [111].
Given the plethora of single cell assays, it is an exciting time for human immunology. However, deciding which assay to use can be challenging and is again dependent on the study goals and questions of interest. Other considerations include cost, these are expensive technologies, and how to analyze the data, cutting-edge bioinformatics expertise is essential to make biological sense of the wealth of data generated. Furthermore, although single cell profiling can be useful for discovery, it should always be complemented with functional assays. This is especially important because studying the immune response to perturbations may reveal changes not seen in the resting state. This was the observed in T1D where the TruCulture whole blood stimulation assay [112] revealed type 1 interferon (IFN-1) hyper-responsiveness that was not detected at baseline [113]. Again, the choice of functional assay and stimuli of interest is dependent on the question to be addressed but the assays can be in vitro or directly ex vivo and can test a range of responses from activation in response to antigen, cytokines, and interactions with ligands such as toll-like receptor (TLR) ligands. Lastly, validation is also a requirement for a well-designed study of human immunology, including independent validation cohorts and lab-based assays assessing loss- and gain-of-function. Further validation can also be achieved by mining publicly available datasets.
Concluding remarks
In the past, human immunology studies were given second class status compared to studies using model systems due to the difficulty in proving mechanism and the variability of data generated from human samples. Today we have the tools to overcome many of these past challenges and can now produce high quality data and manipulate human cells and systems in ways that lead to an understanding of disease mechanisms that was not possible in the past. To continue to advance our understanding of human immunology in the next decade, we will need: 1) long term commitments to funding, 2) collaborations across areas of expertise and institutions, and 3) ability to integrate and analyze large data sets. With this toolkit we can now address the questions most salient to patients and their caregivers. Who is at risk for a disease, how can we prevent it, and, if someone has a newly diagnosed disease, what will be the most effective therapy for them to control or reverse the disease process.
Acknowledgements:
Funding from the National Institutes of Health (R01 AI132774 and U01 AI101990) help support the development of the ideas presented here. I would also like to acknowledge my colleagues at the Benaroya Research Institute at Virginia Mason for their helpful discussions especially Carla Greenbaum and Cate Speake in the Center for Interventional Immunology and Alice Long and Karen Cerosaletti in the Center for Translational Immunology. The Center for Interventional Immunology and the Clinical Core Lab have also provided invaluable support for the BRI Repository and Registry. A special thank you to all the healthy individuals and patients who have participated in the BRI Registry and Repository. Anne Hocking and Taylor Lawson in the BRI Scientific Writing Group assisted with writing and figure preparation.
Abbreviations:
- ACPA
anti-citrullinated protein antibodies
- ASAP-seq
ATAC with select antigen profiling by sequencing
- scATAC-seq
single cell assay for transposase-accessible chromatin using sequencing
- BCR
B cell receptor
- CAR-T
chimeric antigen receptor T cell
- CITE-seq
cellular indexing of transcriptomes and epitopes by sequencing
- CTLA-4
cytotoxic T lymphocyte associated protein 4
- DOGMA-seq
done overfitting genomics methods acronym
- EMU
experimental medicine unit
- eQTL
expression quantitative trait loci
- GWAS
genome-wide association study
- HLA
human leukocyte antigen
- ICICLE-seq
integrated cellular indexing of chromatin landscape and epitopes
- IFN-1
type 1 interferon
- MHC
major histocompatibility complex
- MS
multiple sclerosis
- mtDNA
mitochondrial DNA
- Paired-Tag
parallel analysis of individual cells for RNA expression and DNA from targeted tagmentation by sequencing
- PTPN22
protein tyrosine phosphatase non-receptor type 22
- RA
rheumatoid arthritis
- REAP-seq
RNA expression and protein sequencing
- scCUT&Tag
single cell cleavage under targets and tagmentation
- sci-CAR
single-cell combinatorial indexing chromatin accessibility and mRNA
- scRNA-seq
single cell RNA sequencing
- SHARE-seq
simultaneous high-throughput ATAC and RNA expression with sequencing
- SLE
systemic lupus erythematosus
- SNARE-seq
single-nucleus chromatin accessibility and mRNA expression sequencing
- TCR
T cell receptor
- TLR
toll-like receptor
- TEA-seq
simultaneously measures transcriptomics (scRNA-seq), epitopes and chromatin accessibility (scATAC-seq)
- T1D
type 1 diabetes
- TYK2
tyrosine kinase 2
Footnotes
Conflict of Interest: J.H.B. is a Scientific Co-Founder and Scientific Advisory Board member of GentiBio, a consultant for Bristol-Myers Squibb and Hotspot Therapeutics, and has past and current research projects sponsored by Amgen, Bristol-Myers Squib, Janssen, Novo Nordisk, and Pfizer. She is a member of the Type 1 Diabetes TrialNet Study Group, a partner of the Allen Institute for Immunology, and a member of the Scientific Advisory Boards for the La Jolla Institute for Allergy and Immunology and BMS Immunology.
Data availability statement:
Data sharing is not applicable to this article as no new data were created or analyzed in this study.
References
- 1.Tebani A, Gummesson A, Zhong W, Koistinen IS, Lakshmikanth T, Olsson LM, Boulund F, Neiman M, Stenlund H, Hellström C, Karlsson MJ, Arif M, Dodig-Crnković T, Mardinoglu A, Lee S, Zhang C, Chen Y, Olin A, Mikes J, Danielsson H, von Feilitzen K, Jansson PA, Angerås O, Huss M, Kjellqvist S, Odeberg J, Edfors F, Tremaroli V, Forsström B, Schwenk JM, Nilsson P, Moritz T, Bäckhed F, Engstrand L, Brodin P, Bergström G, Uhlen M and Fagerberg L, Integration of molecular profiles in a longitudinal wellness profiling cohort. Nat Commun 2020. 11: 4487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Roederer M, Quaye L, Mangino M, Beddall MH, Mahnke Y, Chattopadhyay P, Tosi I, Napolitano L, Terranova Barberio M, Menni C, Villanova F, Di Meglio P, Spector TD and Nestle FO, The genetic architecture of the human immune system: a bioresource for autoimmunity and disease pathogenesis. Cell 2015. 161: 387–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Brodin P, Jojic V, Gao T, Bhattacharya S, Angel CJ, Furman D, Shen-Orr S, Dekker CL, Swan GE, Butte AJ, Maecker HT and Davis MM, Variation in the human immune system is largely driven by non-heritable influences. Cell 2015. 160: 37–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Mangino M, Roederer M, Beddall MH, Nestle FO and Spector TD, Innate and adaptive immune traits are differentially affected by genetic and environmental factors. Nat Commun 2017. 8: 13850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kaczorowski KJ, Shekhar K, Nkulikiyimfura D, Dekker CL, Maecker H, Davis MM, Chakraborty AK and Brodin P, Continuous immunotypes describe human immune variation and predict diverse responses. Proc Natl Acad Sci U S A 2017. 114: E6097–e6106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Lee DM and Weinblatt ME, Rheumatoid arthritis. Lancet 2001. 358: 903–911. [DOI] [PubMed] [Google Scholar]
- 7.Sokolova MV, Schett G and Steffen U, Autoantibodies in Rheumatoid Arthritis: Historical Background and Novel Findings. Clin Rev Allergy Immunol 2022. 63: 138–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Nepom GT, Major histocompatibility complex-directed susceptibility to rheumatoid arthritis. Adv Immunol 1998. 68: 315–332. [DOI] [PubMed] [Google Scholar]
- 9.Eyre S, Bowes J, Diogo D, Lee A, Barton A, Martin P, Zhernakova A, Stahl E, Viatte S, McAllister K, Amos CI, Padyukov L, Toes RE, Huizinga TW, Wijmenga C, Trynka G, Franke L, Westra HJ, Alfredsson L, Hu X, Sandor C, de Bakker PI, Davila S, Khor CC, Heng KK, Andrews R, Edkins S, Hunt SE, Langford C, Symmons D, Biologics in Rheumatoid Arthritis, G., Genomics Study, S., Wellcome Trust Case Control, C., Concannon P, Onengut-Gumuscu S, Rich SS, Deloukas P, Gonzalez-Gay MA, Rodriguez-Rodriguez L, Arlsetig L, Martin J, Rantapaa-Dahlqvist S, Plenge RM, Raychaudhuri S, Klareskog L, Gregersen PK and Worthington J, High-density genetic mapping identifies new susceptibility loci for rheumatoid arthritis. Nat Genet 2012. 44: 1336–1340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ishigaki K, Sakaue S, Terao C, Luo Y, Sonehara K, Yamaguchi K, Amariuta T, Too CL, Laufer VA, Scott IC, Viatte S, Takahashi M, Ohmura K, Murasawa A, Hashimoto M, Ito H, Hammoudeh M, Emadi SA, Masri BK, Halabi H, Badsha H, Uthman IW, Wu X, Lin L, Li T, Plant D, Barton A, Orozco G, Verstappen SMM, Bowes J, MacGregor AJ, Honda S, Koido M, Tomizuka K, Kamatani Y, Tanaka H, Tanaka E, Suzuki A, Maeda Y, Yamamoto K, Miyawaki S, Xie G, Zhang J, Amos CI, Keystone E, Wolbink G, van der Horst-Bruinsma I, Cui J, Liao KP, Carroll RJ, Lee HS, Bang SY, Siminovitch KA, de Vries N, Alfredsson L, Rantapää-Dahlqvist S, Karlson EW, Bae SC, Kimberly RP, Edberg JC, Mariette X, Huizinga T, Dieudé P, Schneider M, Kerick M, Denny JC, Matsuda K, Matsuo K, Mimori T, Matsuda F, Fujio K, Tanaka Y, Kumanogoh A, Traylor M, Lewis CM, Eyre S, Xu H, Saxena R, Arayssi T, Kochi Y, Ikari K, Harigai M, Gregersen PK, Yamamoto K, Louis Bridges S Jr., Padyukov L, Martin J, Klareskog L, Okada Y and Raychaudhuri S, Multi-ancestry genome-wide association analyses identify novel genetic mechanisms in rheumatoid arthritis. Nat Genet 2022. 54: 1640–1651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Saevarsdottir S, Stefansdottir L, Sulem P, Thorleifsson G, Ferkingstad E, Rutsdottir G, Glintborg B, Westerlind H, Grondal G, Loft IC, Sorensen SB, Lie BA, Brink M, Ärlestig L, Arnthorsson AO, Baecklund E, Banasik K, Bank S, Bjorkman LI, Ellingsen T, Erikstrup C, Frei O, Gjertsson I, Gudbjartsson DF, Gudjonsson SA, Halldorsson GH, Hendricks O, Hillert J, Hogdall E, Jacobsen S, Jensen DV, Jonsson H, Kastbom A, Kockum I, Kristensen S, Kristjansdottir H, Larsen MH, Linauskas A, Hauge EM, Loft AG, Ludviksson BR, Lund SH, Markusson T, Masson G, Melsted P, Moore KHS, Munk H, Nielsen KR, Norddahl GL, Oddsson A, Olafsdottir TA, Olason PI, Olsson T, Ostrowski SR, Hørslev-Petersen K, Rognvaldsson S, Sanner H, Silberberg GN, Stefansson H, Sørensen E, Sørensen IJ, Turesson C, Bergman T, Alfredsson L, Kvien TK, Brunak S, Steinsson K, Andersen V, Andreassen OA, Rantapää-Dahlqvist S, Hetland ML, Klareskog L, Askling J, Padyukov L, Pedersen OB, Thorsteinsdottir U, Jonsdottir I and Stefansson K, Multiomics analysis of rheumatoid arthritis yields sequence variants that have large effects on risk of the seropositive subset. Ann Rheum Dis 2022. 81: 1085–1095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.MacGregor AJ, Snieder H, Rigby AS, Koskenvuo M, Kaprio J, Aho K and Silman AJ, Characterizing the quantitative genetic contribution to rheumatoid arthritis using data from twins. Arthritis Rheum. 2000. 43: 30–37. [DOI] [PubMed] [Google Scholar]
- 13.van der Woude D, Houwing-Duistermaat JJ, Toes RE, Huizinga TW, Thomson W, Worthington J, van der Helm-van Mil AH and de Vries RR, Quantitative heritability of anti-citrullinated protein antibody-positive and anti-citrullinated protein antibody-negative rheumatoid arthritis. Arthritis Rheum 2009. 60: 916–923. [DOI] [PubMed] [Google Scholar]
- 14.van Delft MAM and Huizinga TWJ, An overview of autoantibodies in rheumatoid arthritis. J Autoimmun 2020. 110: 102392. [DOI] [PubMed] [Google Scholar]
- 15.Silman AJ, Newman J and MacGregor AJ, Cigarette smoking increases the risk of rheumatoid arthritis. Results from a nationwide study of disease-discordant twins. Arthritis Rheum 1996. 39: 732–735. [DOI] [PubMed] [Google Scholar]
- 16.Mahdi H, Fisher BA, Källberg H, Plant D, Malmström V, Rönnelid J, Charles P, Ding B, Alfredsson L, Padyukov L, Symmons DP, Venables PJ, Klareskog L and Lundberg K, Specific interaction between genotype, smoking and autoimmunity to citrullinated alpha-enolase in the etiology of rheumatoid arthritis. Nat Genet 2009. 41: 1319–1324. [DOI] [PubMed] [Google Scholar]
- 17.Chriswell ME, Lefferts AR, Clay MR, Hsu AR, Seifert J, Feser ML, Rims C, Bloom MS, Bemis EA, Liu S, Maerz MD, Frank DN, Demoruelle MK, Deane KD, James EA, Buckner JH, Robinson WH, Holers VM and Kuhn KA, Clonal IgA and IgG autoantibodies from individuals at risk for rheumatoid arthritis identify an arthritogenic strain of Subdoligranulum. Sci Transl Med 2022. 14: eabn5166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Mikuls TR, Thiele GM, Deane KD, Payne JB, O’Dell JR, Yu F, Sayles H, Weisman MH, Gregersen PK, Buckner JH, Keating RM, Derber LA, Robinson WH, Holers VM and Norris JM, Porphyromonas gingivalis and disease-related autoantibodies in individuals at increased risk of rheumatoid arthritis. Arthritis Rheum 2012. 64: 3522–3530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Krutyhołowa A, Strzelec K, Dziedzic A, Bereta GP, Łazarz-Bartyzel K, Potempa J and Gawron K, Host and bacterial factors linking periodontitis and rheumatoid arthritis. Front Immunol 2022. 13: 980805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Maini R, St Clair EW, Breedveld F, Furst D, Kalden J, Weisman M, Smolen J, Emery P, Harriman G, Feldmann M and Lipsky P, Infliximab (chimeric anti-tumour necrosis factor alpha monoclonal antibody) versus placebo in rheumatoid arthritis patients receiving concomitant methotrexate: a randomised phase III trial. ATTRACT Study Group. Lancet 1999. 354: 1932–1939. [DOI] [PubMed] [Google Scholar]
- 21.Lipsky PE, van der Heijde DM, St Clair EW, Furst DE, Breedveld FC, Kalden JR, Smolen JS, Weisman M, Emery P, Feldmann M, Harriman GR and Maini RN, Infliximab and methotrexate in the treatment of rheumatoid arthritis. Anti-Tumor Necrosis Factor Trial in Rheumatoid Arthritis with Concomitant Therapy Study Group. N Engl J Med 2000. 343: 1594–1602. [DOI] [PubMed] [Google Scholar]
- 22.Weinblatt ME, Keystone EC, Furst DE, Moreland LW, Weisman MH, Birbara CA, Teoh LA, Fischkoff SA and Chartash EK, Adalimumab, a fully human anti-tumor necrosis factor alpha monoclonal antibody, for the treatment of rheumatoid arthritis in patients taking concomitant methotrexate: the ARMADA trial. Arthritis Rheum 2003. 48: 35–45. [DOI] [PubMed] [Google Scholar]
- 23.Hetland ML, Christensen IJ, Tarp U, Dreyer L, Hansen A, Hansen IT, Kollerup G, Linde L, Lindegaard HM, Poulsen UE, Schlemmer A, Jensen DV, Jensen S, Hostenkamp G and Østergaard M, Direct comparison of treatment responses, remission rates, and drug adherence in patients with rheumatoid arthritis treated with adalimumab, etanercept, or infliximab: results from eight years of surveillance of clinical practice in the nationwide Danish DANBIO registry. Arthritis Rheum 2010. 62: 22–32. [DOI] [PubMed] [Google Scholar]
- 24.Singh JA, Saag KG, Bridges SL Jr., Akl EA, Bannuru RR, Sullivan MC, Vaysbrot E, McNaughton C, Osani M, Shmerling RH, Curtis JR, Furst DE, Parks D, Kavanaugh A, O’Dell J, King C, Leong A, Matteson EL, Schousboe JT, Drevlow B, Ginsberg S, Grober J, St Clair EW, Tindall E, Miller AS and McAlindon T, 2015 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis. Arthritis Rheumatol 2016. 68: 1–26. [DOI] [PubMed] [Google Scholar]
- 25.Khader Y, Beran A, Ghazaleh S, Lee-Smith W and Altorok N, Predictors of remission in rheumatoid arthritis patients treated with biologics: a systematic review and meta-analysis. Clin Rheumatol 2022. 41: 3615–3627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Multiple sclerosis genomic map implicates peripheral immune cells and microglia in susceptibility. Science 2019. 365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.International Multiple Sclerosis Genetics, C., Beecham AH, Patsopoulos NA, Xifara DK, Davis MF, Kemppinen A, Cotsapas C, Shah TS, Spencer C, Booth D, Goris A, Oturai A, Saarela J, Fontaine B, Hemmer B, Martin C, Zipp F, D’Alfonso S, Martinelli-Boneschi F, Taylor B, Harbo HF, Kockum I, Hillert J, Olsson T, Ban M, Oksenberg JR, Hintzen R, Barcellos LF, Wellcome Trust Case Control, C., International, I. B. D. G. C., Agliardi C, Alfredsson L, Alizadeh M, Anderson C, Andrews R, Sondergaard HB, Baker A, Band G, Baranzini SE, Barizzone N, Barrett J, Bellenguez C, Bergamaschi L, Bernardinelli L, Berthele A, Biberacher V, Binder TM, Blackburn H, Bomfim IL, Brambilla P, Broadley S, Brochet B, Brundin L, Buck D, Butzkueven H, Caillier SJ, Camu W, Carpentier W, Cavalla P, Celius EG, Coman I, Comi G, Corrado L, Cosemans L, Cournu-Rebeix I, Cree BA, Cusi D, Damotte V, Defer G, Delgado SR, Deloukas P, di Sapio A, Dilthey AT, Donnelly P, Dubois B, Duddy M, Edkins S, Elovaara I, Esposito F, Evangelou N, Fiddes B, Field J, Franke A, Freeman C, Frohlich IY, Galimberti D, Gieger C, Gourraud PA, Graetz C, Graham A, Grummel V, Guaschino C, Hadjixenofontos A, Hakonarson H, Halfpenny C, Hall G, Hall P, Hamsten A, Harley J, Harrower T, Analysis of immune-related loci identifies 48 new susceptibility variants for multiple sclerosis. Nat Genet 2013. 45: 1353–1360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Wang YF, Zhang Y, Lin Z, Zhang H, Wang TY, Cao Y, Morris DL, Sheng Y, Yin X, Zhong SL, Gu X, Lei Y, He J, Wu Q, Shen JJ, Yang J, Lam TH, Lin JH, Mai ZM, Guo M, Tang Y, Chen Y, Song Q, Ban B, Mok CC, Cui Y, Lu L, Shen N, Sham PC, Lau CS, Smith DK, Vyse TJ, Zhang X, Lau YL and Yang W, Identification of 38 novel loci for systemic lupus erythematosus and genetic heterogeneity between ancestral groups. Nat Commun 2021. 12: 772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Chen L, Morris DL and Vyse TJ, Genetic advances in systemic lupus erythematosus: an update. Curr Opin Rheumatol 2017. 29: 423–433. [DOI] [PubMed] [Google Scholar]
- 30.Chen L, Wang YF, Liu L, Bielowka A, Ahmed R, Zhang H, Tombleson P, Roberts AL, Odhams CA, Cunninghame Graham DS, Zhang X, Yang W, Vyse TJ and Morris DL, Genome-wide assessment of genetic risk for systemic lupus erythematosus and disease severity. Hum Mol Genet 2020. 29: 1745–1756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Simpson S Jr., Wang W, Otahal P, Blizzard L, van der Mei IAF and Taylor BV, Latitude continues to be significantly associated with the prevalence of multiple sclerosis: an updated meta-analysis. J Neurol Neurosurg Psychiatry 2019. 90: 1193–1200. [DOI] [PubMed] [Google Scholar]
- 32.Woo JMP, Parks CG, Jacobsen S, Costenbader KH and Bernatsky S, The role of environmental exposures and gene-environment interactions in the etiology of systemic lupus erythematous. J Intern Med 2022. 291: 755–778. [DOI] [PubMed] [Google Scholar]
- 33.Bjornevik K, Cortese M, Healy BC, Kuhle J, Mina MJ, Leng Y, Elledge SJ, Niebuhr DW, Scher AI, Munger KL and Ascherio A, Longitudinal analysis reveals high prevalence of Epstein-Barr virus associated with multiple sclerosis. Science 2022. 375: 296–301. [DOI] [PubMed] [Google Scholar]
- 34.Iwata S and Tanaka Y, Association of Viral Infection With the Development and Pathogenesis of Systemic Lupus Erythematosus. Front Med (Lausanne) 2022. 9: 849120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Morand EF, Furie R, Tanaka Y, Bruce IN, Askanase AD, Richez C, Bae SC, Brohawn PZ, Pineda L, Berglind A and Tummala R, Trial of Anifrolumab in Active Systemic Lupus Erythematosus. N Engl J Med 2020. 382: 211–221. [DOI] [PubMed] [Google Scholar]
- 36.Wyse C, O’Malley G, Coogan AN, McConkey S and Smith DJ, Seasonal and daytime variation in multiple immune parameters in humans: Evidence from 329,261 participants of the UK Biobank cohort. iScience 2021. 24: 102255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Wang C, Lutes LK, Barnoud C and Scheiermann C, The circadian immune system. Sci Immunol 2022. 7: eabm2465. [DOI] [PubMed] [Google Scholar]
- 38.Cermakian N and Labrecque N, Regulation of Cytotoxic CD8+ T Cells by the Circadian Clock. J Immunol 2023. 210: 12–18. [DOI] [PubMed] [Google Scholar]
- 39.Christ A, Lauterbach M and Latz E, Western Diet and the Immune System: An Inflammatory Connection. Immunity 2019. 51: 794–811. [DOI] [PubMed] [Google Scholar]
- 40.Dopico XC, Evangelou M, Ferreira RC, Guo H, Pekalski ML, Smyth DJ, Cooper N, Burren OS, Fulford AJ, Hennig BJ, Prentice AM, Ziegler AG, Bonifacio E, Wallace C and Todd JA, Widespread seasonal gene expression reveals annual differences in human immunity and physiology. Nat Commun 2015. 6: 7000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lange T, Dimitrov S, Bollinger T, Diekelmann S and Born J, Sleep after vaccination boosts immunological memory. J Immunol 2011. 187: 283–290. [DOI] [PubMed] [Google Scholar]
- 42.Azad MB, Lissitsyn Y, Miller GE, Becker AB, HayGlass KT and Kozyrskyj AL, Influence of socioeconomic status trajectories on innate immune responsiveness in children. PLoS One 2012. 7: e38669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Dhabhar FS, Miller AH, McEwen BS and Spencer RL, Effects of stress on immune cell distribution. Dynamics and hormonal mechanisms. J Immunol 1995. 154: 5511–5527. [PubMed] [Google Scholar]
- 44.Shirai Y, Nakanishi Y, Suzuki A, Konaka H, Nishikawa R, Sonehara K, Namba S, Tanaka H, Masuda T, Yaga M, Satoh S, Izumi M, Mizuno Y, Jo T, Maeda Y, Nii T, Oguro-Igashira E, Morisaki T, Kamatani Y, Nakayamada S, Nishigori C, Tanaka Y, Takeda Y, Yamamoto K, Kumanogoh A and Okada Y, Multi-trait and cross-population genome-wide association studies across autoimmune and allergic diseases identify shared and distinct genetic component. Ann Rheum Dis 2022. 81: 1301–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Saurabh R, Fouodo CJK, König IR, Busch H and Wohlers I, A survey of genome-wide association studies, polygenic scores and UK Biobank highlights resources for autoimmune disease genetics. Front Immunol 2022. 13: 972107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Mahon JL, Sosenko JM, Rafkin-Mervis L, Krause-Steinrauf H, Lachin JM, Thompson C, Bingley PJ, Bonifacio E, Palmer JP, Eisenbarth GS, Wolfsdorf J and Skyler JS, The TrialNet Natural History Study of the Development of Type 1 Diabetes: objectives, design, and initial results. Pediatr.Diabetes 2009. 10: 97–104. [DOI] [PubMed] [Google Scholar]
- 47.Herold KC, Usmani-Brown S, Ghazi T, Lebastchi J, Beam CA, Bellin MD, Ledizet M, Sosenko JM, Krischer JP, Palmer JP and Type 1 Diabetes TrialNet Study, G., beta cell death and dysfunction during type 1 diabetes development in at-risk individuals. J Clin Invest 2015. 125: 1163–1173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.TEDDY Study Group, The Environmental Determinants of Diabetes in the Young (TEDDY) Study. Ann N Y Acad Sci 2008. 1150: 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Roll U, Christie MR, Füchtenbusch M, Payton MA, Hawkes CJ and Ziegler AG, Perinatal autoimmunity in offspring of diabetic parents. The German Multicenter BABY-DIAB study: detection of humoral immune responses to islet antigens in early childhood. Diabetes 1996. 45: 967–973. [DOI] [PubMed] [Google Scholar]
- 50.Ziegler AG, Hummel M, Schenker M and Bonifacio E, Autoantibody appearance and risk for development of childhood diabetes in offspring of parents with type 1 diabetes: the 2-year analysis of the German BABYDIAB Study. Diabetes 1999. 48: 460–468. [DOI] [PubMed] [Google Scholar]
- 51.Ziegler AG, Rewers M, Simell O, Simell T, Lempainen J, Steck A, Winkler C, Ilonen J, Veijola R, Knip M, Bonifacio E and Eisenbarth GS, Seroconversion to multiple islet autoantibodies and risk of progression to diabetes in children. Jama 2013. 309: 2473–2479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Insel RA, Dunne JL, Atkinson MA, Chiang JL, Dabelea D, Gottlieb PA, Greenbaum CJ, Herold KC, Krischer JP, Lernmark A, Ratner RE, Rewers MJ, Schatz DA, Skyler JS, Sosenko JM and Ziegler AG, Staging presymptomatic type 1 diabetes: a scientific statement of JDRF, the Endocrine Society, and the American Diabetes Association. Diabetes Care 2015. 38: 1964–1974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Herold KC, Bundy BN, Long SA, Bluestone JA, DiMeglio LA, Dufort MJ, Gitelman SE, Gottlieb PA, Krischer JP, Linsley PS, Marks JB, Moore W, Moran A, Rodriguez H, Russell WE, Schatz D, Skyler JS, Tsalikian E, Wherrett DK, Ziegler AG, Greenbaum CJ and Type 1 Diabetes TrialNet Study, G., An Anti-CD3 Antibody, Teplizumab, in Relatives at Risk for Type 1 Diabetes. N Engl J Med 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kolfenbach JR, Deane KD, Derber LA, O’Donnell C, Weisman MH, Buckner JH, Gersuk VH, Wei S, Mikuls TR, O’Dell J, Gregersen PK, Keating RM, Norris JM and Holers VM, A prospective approach to investigating the natural history of preclinical rheumatoid arthritis (RA) using first-degree relatives of probands with RA. Arthritis Rheum 2009. 61: 1735–1742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Gilbert BTP, Lamacchia C, Mongin D, Lauper K, Trunk E, Studer O, Courvoisier DS, Rubbert-Roth A, Kyburz D, Moeller B and Finckh A, Cohort profile: SCREEN-RA: design, methods and perspectives of a Swiss cohort study of first-degree relatives of patients with rheumatoid arthritis. BMJ Open 2021. 11: e048409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Bhattacharya J, Pappas K, Toz B, Aranow C, Mackay M, Gregersen PK, Doumbo O, Traore AK, Lesser ML, McMahon M, Utset T, Silverman E, Levy D, McCune WJ, Jolly M, Wallace D, Weisman M, Romero-Diaz J and Diamond B, Serologic features of cohorts with variable genetic risk for systemic lupus erythematosus. Mol Med 2018. 24: 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Redmond C, Pamuk O and Hasni SA, Lupus Cohorts. Rheum Dis Clin North Am 2021. 47: 457–479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Taylor KM, Hanscombe KB, Prescott NJ, Iniesta R, Traylor M, Taylor NS, Fong S, Powell N, Irving PM, Anderson SH, Mathew CG, Lewis CM and Sanderson JD, Genetic and Inflammatory Biomarkers Classify Small Intestine Inflammation in Asymptomatic First-degree Relatives of Patients With Crohn’s Disease. Clin Gastroenterol Hepatol 2020. 18: 908–916.e913. [DOI] [PubMed] [Google Scholar]
- 59.Finckh A, Gilbert B, Hodkinson B, Bae SC, Thomas R, Deane KD, Alpizar-Rodriguez D and Lauper K, Global epidemiology of rheumatoid arthritis. Nat Rev Rheumatol 2022. 18: 591–602. [DOI] [PubMed] [Google Scholar]
- 60.Drenkard C and Lim SS, Update on lupus epidemiology: advancing health disparities research through the study of minority populations. Curr Opin Rheumatol 2019. 31: 689–696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Langer-Gould AM, Gonzales EG, Smith JB, Li BH and Nelson LM, Racial and Ethnic Disparities in Multiple Sclerosis Prevalence. Neurology 2022. 98: e1818–e1827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Szabo PA, Miron M and Farber DL, Location, location, location: Tissue resident memory T cells in mice and humans. Sci Immunol 2019. 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Aida K, Fukui T, Jimbo E, Yagihashi S, Shimada A, Oikawa Y, Mori Y, Fujii T, Nishida Y, Koyama R and Kobayashi T, Distinct Inflammatory Changes of the Pancreas of Slowly Progressive Insulin-dependent (Type 1) Diabetes. Pancreas 2018. 47: 1101–1109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Richardson SJ and Pugliese A, 100 YEARS OF INSULIN: Pancreas pathology in type 1 diabetes: an evolving story. J Endocrinol 2021. 252: R41–r57. [DOI] [PubMed] [Google Scholar]
- 65.Richardson SJ, Morgan NG and Foulis AK, Pancreatic pathology in type 1 diabetes mellitus. Endocr Pathol 2014. 25: 80–92. [DOI] [PubMed] [Google Scholar]
- 66.Zhang F, Wei K, Slowikowski K, Fonseka CY, Rao DA, Kelly S, Goodman SM, Tabechian D, Hughes LB, Salomon-Escoto K, Watts GFM, Jonsson AH, Rangel-Moreno J, Meednu N, Rozo C, Apruzzese W, Eisenhaure TM, Lieb DJ, Boyle DL, Mandelin AM 2nd, Boyce BF, DiCarlo E, Gravallese EM, Gregersen PK, Moreland L, Firestein GS, Hacohen N, Nusbaum C, Lederer JA, Perlman H, Pitzalis C, Filer A, Holers VM, Bykerk VP, Donlin LT, Anolik JH, Brenner MB and Raychaudhuri S, Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nat Immunol 2019. 20: 928–942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.The “All of Us” Research Program. New England Journal of Medicine 2019. 381: 668–676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Thomas S, Rouilly V, Patin E, Alanio C, Dubois A, Delval C, Marquier LG, Fauchoux N, Sayegrih S, Vray M, Duffy D, Quintana-Murci L and Albert ML, The Milieu Intérieur study - an integrative approach for study of human immunological variance. Clin Immunol 2015. 157: 277–293. [DOI] [PubMed] [Google Scholar]
- 69.Long SA, Thorpe J, DeBerg HA, Gersuk V, Eddy J, Harris KM, Ehlers M, Herold KC, Nepom GT and Linsley PS, Partial exhaustion of CD8 T cells and clinical response to teplizumab in new-onset type 1 diabetes. Sci Immunol 2016. 1: 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Habib T, Long SA, Samuels PL, Brahmandam A, Tatum M, Funk A, Hocking AM, Cerosaletti K, Mason MT, Whalen E, Rawlings DJ, Greenbaum C, Buckner JH and Type 1 Diabetes TrialNet Study, G., Dynamic Immune Phenotypes of B and T Helper Cells Mark Distinct Stages of T1D Progression. Diabetes 2019. 68: 1240–1250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Linsley PS, Greenbaum CJ, Rosasco M, Presnell S, Herold KC and Dufort MJ, Elevated T cell levels in peripheral blood predict poor clinical response following rituximab treatment in new-onset type 1 diabetes. Genes Immun 2018. 20: 293–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Sanda S, Bollyky J, Standifer N, Nepom G, Hamerman JA and Greenbaum C, Short-term IL-1beta blockade reduces monocyte CD11b integrin expression in an IL-8 dependent fashion in patients with type 1 diabetes. Clin Immunol 2010. 136: 170–173. [DOI] [PubMed] [Google Scholar]
- 73.Bollyky JB, Long SA, Fitch M, Bollyky PL, Rieck M, Rogers R, Samuels PL, Sanda S, Buckner JH, Hellerstein MK and Greenbaum CJ, Evaluation of in vivo T cell kinetics: use of heavy isotope labelling in type 1 diabetes. Clinical and Experimental Immunology 2013. 172: 363–374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Speake C, Habib T, Lambert K, Hundhausen C, Lord S, Dufort MJ, Skinner SO, Hu A, Kinsman M, Jones BE, Maerz MD, Tatum M, Hocking AM, Nepom GT, Greenbaum CJ and Buckner JH, IL-6-targeted therapies to block the cytokine or its receptor drive distinct alterations in T cell function. JCI Insight 2022. 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Stephenson E, Webb S and Haniffa M, Multiomics uncovers developing immunological lineages in human. Eur J Immunol 2021. 51: 764–772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Xu Z, Heidrich-O’Hare E, Chen W and Duerr RH, Comprehensive benchmarking of CITE-seq versus DOGMA-seq single cell multimodal omics. Genome Biol 2022. 23: 135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Stoeckius M, Hafemeister C, Stephenson W, Houck-Loomis B, Chattopadhyay PK, Swerdlow H, Satija R and Smibert P, Simultaneous epitope and transcriptome measurement in single cells. Nat Methods 2017. 14: 865–868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Peterson VM, Zhang KX, Kumar N, Wong J, Li L, Wilson DC, Moore R, McClanahan TK, Sadekova S and Klappenbach JA, Multiplexed quantification of proteins and transcripts in single cells. Nat Biotechnol 2017. 35: 936–939. [DOI] [PubMed] [Google Scholar]
- 79.Cao J, Cusanovich DA, Ramani V, Aghamirzaie D, Pliner HA, Hill AJ, Daza RM, McFaline-Figueroa JL, Packer JS, Christiansen L, Steemers FJ, Adey AC, Trapnell C and Shendure J, Joint profiling of chromatin accessibility and gene expression in thousands of single cells. Science 2018. 361: 1380–1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Chen S, Lake BB and Zhang K, High-throughput sequencing of the transcriptome and chromatin accessibility in the same cell. Nat Biotechnol 2019. 37: 1452–1457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ma S, Zhang B, LaFave LM, Earl AS, Chiang Z, Hu Y, Ding J, Brack A, Kartha VK, Tay T, Law T, Lareau C, Hsu YC, Regev A and Buenrostro JD, Chromatin Potential Identified by Shared Single-Cell Profiling of RNA and Chromatin. Cell 2020. 183: 1103–1116.e1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Mimitou EP, Lareau CA, Chen KY, Zorzetto-Fernandes AL, Hao Y, Takeshima Y, Luo W, Huang TS, Yeung BZ, Papalexi E, Thakore PI, Kibayashi T, Wing JB, Hata M, Satija R, Nazor KL, Sakaguchi S, Ludwig LS, Sankaran VG, Regev A and Smibert P, Scalable, multimodal profiling of chromatin accessibility, gene expression and protein levels in single cells. Nat Biotechnol 2021. 39: 1246–1258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Swanson E, Lord C, Reading J, Heubeck AT, Genge PC, Thomson Z, Weiss MD, Li XJ, Savage AK, Green RR, Torgerson TR, Bumol TF, Graybuck LT and Skene PJ, Simultaneous trimodal single-cell measurement of transcripts, epitopes, and chromatin accessibility using TEA-seq. Elife 2021. 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Zhang B, Srivastava A, Mimitou E, Stuart T, Raimondi I, Hao Y, Smibert P and Satija R, Characterizing cellular heterogeneity in chromatin state with scCUT&Tag-pro. Nat Biotechnol 2022. 40: 1220–1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Zhu C, Zhang Y, Li YE, Lucero J, Behrens MM and Ren B, Joint profiling of histone modifications and transcriptome in single cells from mouse brain. Nat Methods 2021. 18: 283–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Yazar S, Alquicira-Hernandez J, Wing K, Senabouth A, Gordon MG, Andersen S, Lu Q, Rowson A, Taylor TRP, Clarke L, Maccora K, Chen C, Cook AL, Ye CJ, Fairfax KA, Hewitt AW and Powell JE, Single-cell eQTL mapping identifies cell type-specific genetic control of autoimmune disease. Science 2022. 376: eabf3041. [DOI] [PubMed] [Google Scholar]
- 87.Perez RK, Gordon MG, Subramaniam M, Kim MC, Hartoularos GC, Targ S, Sun Y, Ogorodnikov A, Bueno R, Lu A, Thompson M, Rappoport N, Dahl A, Lanata CM, Matloubian M, Maliskova L, Kwek SS, Li T, Slyper M, Waldman J, Dionne D, Rozenblatt-Rosen O, Fong L, Dall’Era M, Balliu B, Regev A, Yazdany J, Criswell LA, Zaitlen N and Ye CJ, Single-cell RNA-seq reveals cell type-specific molecular and genetic associations to lupus. Science 2022. 376: eabf1970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Orange DE, Yao V, Sawicka K, Fak J, Frank MO, Parveen S, Blachere NE, Hale C, Zhang F, Raychaudhuri S, Troyanskaya OG and Darnell RB, RNA Identification of PRIME Cells Predicting Rheumatoid Arthritis Flares. N Engl J Med 2020. 383: 218–228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Kotliarov Y, Sparks R, Martins AJ, Mulè MP, Lu Y, Goswami M, Kardava L, Banchereau R, Pascual V, Biancotto A, Chen J, Schwartzberg PL, Bansal N, Liu CC, Cheung F, Moir S and Tsang JS, Broad immune activation underlies shared set point signatures for vaccine responsiveness in healthy individuals and disease activity in patients with lupus. Nat Med 2020. 26: 618–629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.King HW, Wells KL, Shipony Z, Kathiria AS, Wagar LE, Lareau C, Orban N, Capasso R, Davis MM, Steinmetz LM, James LK and Greenleaf WJ, Integrated single-cell transcriptomics and epigenomics reveals strong germinal center-associated etiology of autoimmune risk loci. Sci Immunol 2021. 6: eabh3768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Christophersen A, Peptide-MHC class I and class II tetramers: From flow to mass cytometry. Hla 2020. 95: 169–178. [DOI] [PubMed] [Google Scholar]
- 92.Kwok WW, Ptacek NA, Liu AW and Buckner JH, Use of class II tetramers for identification of CD4+ T cells. J.Immunol.Methods 2002. 268: 71–81. [DOI] [PubMed] [Google Scholar]
- 93.Nepom GT, Buckner JH, Novak EJ, Reichstetter S, Reijonen H, Gebe J, Wang R, Swanson E and Kwok WW, HLA class II tetramers: tools for direct analysis of antigen-specific CD4+ T cells. Arthritis Rheum 2002. 46: 5–12. [DOI] [PubMed] [Google Scholar]
- 94.Cerosaletti K, Barahmand-Pour-Whitman F, Yang J, DeBerg HA, Dufort MJ, Murray SA, Israelsson E, Speake C, Gersuk VH, Eddy JA, Reijonen H, Greenbaum CJ, Kwok WW, Wambre E, Prlic M, Gottardo R, Nepom GT and Linsley PS, Single-Cell RNA Sequencing Reveals Expanded Clones of Islet Antigen-Reactive CD4(+) T Cells in Peripheral Blood of Subjects with Type 1 Diabetes. J Immunol 2017. 199: 323–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Chattopadhyay PK, Yu J and Roederer M, A live-cell assay to detect antigen-specific CD4+ T cells with diverse cytokine profiles. Nature Medicine 2005. 11: 1113–1117. [DOI] [PubMed] [Google Scholar]
- 96.Lu DR, McDavid AN, Kongpachith S, Lingampalli N, Glanville J, Ju CH, Gottardo R and Robinson WH, T Cell-Dependent Affinity Maturation and Innate Immune Pathways Differentially Drive Autoreactive B Cell Responses in Rheumatoid Arthritis. Arthritis Rheumatol 2018. 70: 1732–1744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Boonyaratanakornkit J and Taylor JJ, Techniques to Study Antigen-Specific B Cell Responses. Front Immunol 2019. 10: 1694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Linsley PS, Barahmand-Pour-Whitman F, Balmas E, DeBerg HA, Flynn KJ, Hu AK, Rosasco MG, Chen J, O’Rourke C, Serti E, Gersuk VH, Motwani K, Seay HR, Brusko TM, Kwok WW, Speake C, Greenbaum CJ, Nepom GT and Cerosaletti K, Autoreactive T cell receptors with shared germline-like α chains in type 1 diabetes. JCI Insight 2021. 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Lanz TV, Brewer RC, Ho PP, Moon JS, Jude KM, Fernandez D, Fernandes RA, Gomez AM, Nadj GS, Bartley CM, Schubert RD, Hawes IA, Vazquez SE, Iyer M, Zuchero JB, Teegen B, Dunn JE, Lock CB, Kipp LB, Cotham VC, Ueberheide BM, Aftab BT, Anderson MS, DeRisi JL, Wilson MR, Bashford-Rogers RJM, Platten M, Garcia KC, Steinman L and Robinson WH, Clonally expanded B cells in multiple sclerosis bind EBV EBNA1 and GlialCAM. Nature 2022. 603: 321–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Ahmed R, Omidian Z, Giwa A, Cornwell B, Majety N, Bell DR, Lee S, Zhang H, Michels A, Desiderio S, Sadegh-Nasseri S, Rabb H, Gritsch S, Suva ML, Cahan P, Zhou R, Jie C, Donner T and Hamad ARA, A Public BCR Present in a Unique Dual-Receptor-Expressing Lymphocyte from Type 1 Diabetes Patients Encodes a Potent T Cell Autoantigen. Cell 2019. 177: 1583–1599.e1516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Hanna SJ, Tatovic D, Thayer TC and Dayan CM, Insights From Single Cell RNA Sequencing Into the Immunology of Type 1 Diabetes- Cell Phenotypes and Antigen Specificity. Front Immunol 2021. 12: 751701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Culina S, Lalanne AI, Afonso G, Cerosaletti K, Pinto S, Sebastiani G, Kuranda K, Nigi L, Eugster A, Osterbye T, Maugein A, McLaren JE, Ladell K, Larger E, Beressi JP, Lissina A, Appay V, Davidson HW, Buus S, Price DA, Kuhn M, Bonifacio E, Battaglia M, Caillat-Zucman S, Dotta F, Scharfmann R, Kyewski B, Mallone R, ImMaDiab Study G, Carel JC, Tubiana-Rufi N, Martinerie L, Poidvin A, JacqzAigrain E, Corvez L, Berruer V, Gautier JF, Baz B, Haddadi N, Andreelli F, Amouyal C, Jaqueminet S, Bourron O, Dasque E, Hartemann A, Lemoine-Yazigi A, Dubois-Laforgue D, Travert F, Feron M, Rolland P, Vignali V, Marre M, Chanson P, Briet C, Guillausseau PJ, Ait-Bachir L, Collet C, Beziaud F, Desforges-Bullet V, Petit-Aubert G, Christin-Maitre S, Feve B, Vatier C, Bourcigaux N, Lautridou C, Lahlou N, Bakouboula P, Elie C, Morel H, Treluyer JM, Gagnerault MC, Maillard C and Jones A, Islet-reactive CD8(+) T cell frequencies in the pancreas, but not in blood, distinguish type 1 diabetic patients from healthy donors. Sci Immunol 2018. 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Lauwerys L, Smits E, Van den Wyngaert T and Elvas F, Radionuclide Imaging of Cytotoxic Immune Cell Responses to Anti-Cancer Immunotherapy. Biomedicines 2022. 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Lee HW, Gangadaran P, Kalimuthu S and Ahn BC, Advances in Molecular Imaging Strategies for In Vivo Tracking of Immune Cells. Biomed Res Int 2016. 2016: 1946585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Tay ZW, Chandrasekharan P, Fellows BD, Arrizabalaga IR, Yu E, Olivo M and Conolly SM, Magnetic Particle Imaging: An Emerging Modality with Prospects in Diagnosis, Targeting and Therapy of Cancer. Cancers (Basel) 2021. 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Put S, Schoonooghe S, Devoogdt N, Schurgers E, Avau A, Mitera T, D’Huyvetter M, De Baetselier P, Raes G, Lahoutte T and Matthys P, SPECT imaging of joint inflammation with Nanobodies targeting the macrophage mannose receptor in a mouse model for rheumatoid arthritis. J Nucl Med 2013. 54: 807–814. [DOI] [PubMed] [Google Scholar]
- 107.Zheng F, Perlman H, Matthys P, Wen Y, Lahoutte T, Muyldermans S, Lu S, De Baetselier P, Schoonooghe S, Devoogdt N and Raes G, Specificity Evaluation and Disease Monitoring in Arthritis Imaging with Complement Receptor of the Ig superfamily targeting Nanobodies. Sci Rep 2016. 6: 35966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Vickovic S, Schapiro D, Carlberg K, Lötstedt B, Larsson L, Hildebrandt F, Korotkova M, Hensvold AH, Catrina AI, Sorger PK, Malmström V, Regev A and Ståhl PL, Three-dimensional spatial transcriptomics uncovers cell type localizations in the human rheumatoid arthritis synovium. Commun Biol 2022. 5: 129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Hardt U, Carlberg K, Af Klint E, Sahlström P, Larsson L, van Vollenhoven A, Hernandez Machado S, Israelsson L, Amara K, Chemin K, Korotkova M, Karlsson Hedestam GB, Catrina AI, Teichmann SA, Ståhl PL and Malmström V, Integrated single cell and spatial transcriptomics reveal autoreactive differentiated B cells in joints of early rheumatoid arthritis. Sci Rep 2022. 12: 11876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Alivernini S, MacDonald L, Elmesmari A, Finlay S, Tolusso B, Gigante MR, Petricca L, Di Mario C, Bui L, Perniola S, Attar M, Gessi M, Fedele AL, Chilaka S, Somma D, Sansom SN, Filer A, McSharry C, Millar NL, Kirschner K, Nerviani A, Lewis MJ, Pitzalis C, Clark AR, Ferraccioli G, Udalova I, Buckley CD, Gremese E, McInnes IB, Otto TD and Kurowska-Stolarska M, Distinct synovial tissue macrophage subsets regulate inflammation and remission in rheumatoid arthritis. Nat Med 2020. 26: 1295–1306. [DOI] [PubMed] [Google Scholar]
- 111.Kaufmann M, Schaupp AL, Sun R, Coscia F, Dendrou CA, Cortes A, Kaur G, Evans HG, Mollbrink A, Navarro JF, Sonner JK, Mayer C, DeLuca GC, Lundeberg J, Matthews PM, Attfield KE, Friese MA, Mann M and Fugger L, Identification of early neurodegenerative pathways in progressive multiple sclerosis. Nat Neurosci 2022. 25: 944–955. [DOI] [PubMed] [Google Scholar]
- 112.Duffy D, Rouilly V, Libri V, Hasan M, Beitz B, David M, Urrutia A, Bisiaux A, Labrie ST, Dubois A, Boneca IG, Delval C, Thomas S, Rogge L, Schmolz M, Quintana-Murci L and Albert ML, Functional analysis via standardized whole-blood stimulation systems defines the boundaries of a healthy immune response to complex stimuli. Immunity 2014. 40: 436–450. [DOI] [PubMed] [Google Scholar]
- 113.Rodrigues KB, Dufort MJ, Llibre A, Speake C, Rahman MJ, Bondet V, Quiel J, Linsley PS, Greenbaum CJ, Duffy D and Tarbell KV, Innate immune stimulation of whole blood reveals IFN-1 hyper-responsiveness in type 1 diabetes. Diabetologia 2020. 63: 1576–1587. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article as no new data were created or analyzed in this study.