Journal Pre-proof The nuts and bolts of omics for the clinical allergist Yamini V. Virkud, M.D., Rachel S. Kelly, Ph.D., Caleb Wood, Jessica A. Lasky-Su, Sc.D. PII:
S1081-1206(19)31195-0
DOI:
https://doi.org/10.1016/j.anai.2019.09.017
Reference:
ANAI 3030
To appear in:
Annals of Allergy, Asthma and Immunology
Received Date: 15 August 2019 Revised Date:
17 September 2019
Accepted Date: 17 September 2019
Please cite this article as: Virkud YV, Kelly RS, Wood C, Lasky-Su JA, The nuts and bolts of omics for the clinical allergist, Annals of Allergy, Asthma and Immunology (2019), doi: https://doi.org/10.1016/ j.anai.2019.09.017. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2019 American College of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved.
PTSIII Order
COVERSHEET Date:
19 September 2019 (18:42:12)
Purchase order number: Not Applicable Due date:
19 September 2019
Production site:
ESI - Elsevier NAHS Journal Production
OPCO:
OCUS
Stage:
S100
Supplier (typesetter):
TnQ Books and Journals Pvt Ltd
Item:
ANAI 3030
PII:
S1081-1206(19)31195-0
Nr of ms pages:
1
DOI:
10.1016/j.anai.2019.09.017
Nr of figures:
3
Nr of B/W figures:
3
Nr of printed colour figures:
0
Production type as sent:
Nr of web colour figures:
0
Version nr:
Nr of e-components:
0 0 batch members present
Article number: PIT:
REV
Language:
English
Batch:
CrossMark:
no
Refers to:
Copyright status:
001
Employer: Copyright received date:
E submission item nr:
ANAI_19-08-0404
License:
EO item nr:
19-08-0404
Embargo:
Editor:
Prof. G.D. Marshall
Embargo until stage:
Received date:
15 August 2019
Revised date:
17 September 2019
Acceptance date:
17 September 2019
Right-hand start:
N
Online publication date: Online version:
CPC:
N
Corrections:
print
In scope for CPC:
N
Corrections-uri:
bam:stage:
Revised proof required: Offprint payment:
N/A
Item title:
The nuts and bolts of omics for the clinical allergist
Item group: Item group description: Section: Dochead: Physical figures: E-components: Funding: First author:
Dr. Yamini V. Virkud
Corresponding author:
Dr. Jessica A. Lasky-Su Brigham and Women's Hospital Medicine 181 Longwood Ave
https://ppts-suppliers.elsevier.com/PtsXsqlPages/ecoversheet.xsql?conn=ppts&itm_id=7527878[9/19/2019 1:42:27 PM]
PTSIII Order
Boston, MA 02115 United States Phone: 6178759992 E-mail:
[email protected]
Item remarks: Type: Editorial Remark: Scheduled for the Dec-19 issue and will be published with an editorial from Wayne Shreffler. lmm Type: Editorial Remark: Corresponding Author email address(es):
[email protected]
XML code
+
https://ppts-suppliers.elsevier.com/PtsXsqlPages/ecoversheet.xsql?conn=ppts&itm_id=7527878[9/19/2019 1:42:27 PM]
19-08-0404R1
Objective: Omics, aka multi-omics, is an emerging area of research that is advancing the use of personalized medicine in clinical practice and is therefore relevant for the practicing allergist. Data Sources: We performed a literature search of a selection of scientific findings in omics and allergy, including variants that may be important to allergy outcomes in the genome, transcriptome, metabolome, microbiome, epigenome, and exposome, among others. Study Selections: Basic science papers and review articles. Results: The use of multi-omic data in clinical practice is changing how clinicians treat their patients whereby more personalized approaches are becoming standard in medical practice and has the potential to transform the treatment of allergies. Conclusion: Multi-omic data are relevant and will become increasingly important for the clinical allergist.
1 1
Introduction: What is “omics”?
2
The term “omics” (aka multi-omics) refers to the usage of high-throughput technology to
3
characterize a set of molecules relevant to a field of biology, and has grown to encompass a
4
variety of “-omes” including the genome (DNA), transcriptome (RNA), proteome (proteins),
5
metabolome (metabolites), microbiome (microbiota), exposome (exposures), and many
6
others.1 In 1957, scientists discovered the central dogma of molecular biology that describes a
7
process whereby DNA in a cell is translated into RNA, which then transfers the information to
8
cellular organelles in order to make proteins essential for all cellular structure and function
9
[Figure 1].2, 3 Accordingly, most of the early scientific focus was on characterizing the genome,
10
transcriptome, and proteome. However, now there are 6 current large omic fields that are
11
being studied extensively today: genome (DNA), transcriptome (RNA), proteome (proteins),
12
epigenome (modification to DNA that alter expression), metabolome (metabolites),
13
microbiome (microbiota), exposome (exposures) [Figure 2].
14
“Systems biology” and “integrative omics” are two terms that refer to use of advanced
15
bioinformatics and sophisticated computational approaches to study interactions between
16
these -omes in explaining the pathophysiology of an organism, also known as the “interactome”
17
4
18
“Big Data”, it has only been in the last decade that technological advances have allowed for
19
rapid sequencing and characterization of molecules that previously took years to perform 1.
20
These improvements in efficiency and cost of sequencing omics datasets, paralleled with
21
development of advanced analytic techniques to process these complex datasets, have led to
22
increased study of omics in a variety of human diseases, including asthma, allergic rhinitis, food
. Because omics generally involves the use of very large data sets of information, often called
2 23
allergies, and immunodeficiencies. As scientists learn more about how changes in the
24
interactome predict outcomes in allergic and immunologic diseases, allergists/immunologists
25
may find themselves using omics technologies to help guide diagnosis and treatment of
26
individual patients, a field known as personalized medicine. We are already beginning to see
27
the first steps of personalized medicine in our field, as clinical immunologists begin to use
28
techniques such as whole exome sequencing, to better define the pathophysiology of primary
29
immunodeficiencies 5.
30
Personalized medicine in allergy/immunology
31
Personalized medicine (aka precision medicine) is a promising approach to studying disease
32
etiology, pathogenesis, progression and treatment, in order to obtain information that is
33
specific to an individual that can be utilized to identify the optimal treatment option for that
34
individual.6 Current diagnostic practices for allergists/immunologists include the use of skin
35
prick and intradermal testing, IgE measurements, pulmonary function testing, and a variety of
36
immunological testing (immunoglobulin levels, flow cytometry, tryptase, complement levels,
37
etc). However, many of these tests, particularly skin tests and IgE levels, are limited in their
38
diagnostic specificity and sensitivity for allergic diseases, and even further limited in their ability
39
to predict disease severity and/or treatment response.7 For example, two patients may both
40
have a positive wheal upon skin testing to peanut, yet one may develop an IgE mediated
41
allergic reaction upon peanut consumption while the other might not. Furthermore, the size of
42
the skin test or peanut-specific IgE level cannot sufficiently inform the patient about their
43
sensitivity to peanut or the severity of reaction they may experience. Scientific research is
44
currently focusing on studying various diseases to determine information that may be relevant
3 45
to inform these personalized approaches to treatment. One of the most promising sources of
46
information to be utilized for personalized approaches to many different disease outcomes,
47
including asthma and allergies, are multi-omics data that can now be generated in real time in a
48
timeframe and at a cost practical for clinical use. By researching these novel omics, scientists
49
may be able to identify factors in one’s specific chemical makeup to predict an individual risk
50
for the development of a disease and response to treatment. 1, 6
51
Below we review the omics data types that are commonly generated today and the clinical and
52
translational impact they may have on the development of personalized medicine.
53
Genomics
54
Of the omic fields, genomics, the comprehensive study of genetic variants, is the most
55
advanced, likely due to early focus on this field in the Human Genome Project.1 The Human
56
Genome Project was an effort started in 1990 to sequence the entirety of several volunteers’
57
DNA and was completed in April 2003.8 It should be noted, however, that the terminology “the
58
human genome” fails to capture the vast differences in individual genomes, due to base pair
59
nucleotide differences known as single nucleotide polymorphisms (SNPs). SNPs account for the
60
unique nature of individual’s DNA, with an average of 20 million differences out of 3.2 billion
61
base pairs, and thus, to explain the variety of manifestations of human disease, each person
62
must be sequenced individually.9 Based on early sequencing methods, this would not have been
63
feasible, as the Human Genome Project took 13 years and cost $2.7 billion dollars to sequence
64
a small number of genomes. In contrast with older techniques, “next generation sequencing”
65
(NGS) refers to a variety of modern technologies used to massively increase the speed,
66
efficiency, and accuracy of sequencing. With the development of NGS, an individual’s genome
4 67
can be sequenced in a few hours for less than $1000, and for several hundred dollars for a
68
limited sequencing of the coding DNA sequences, known as whole exome sequencing.8 The
69
speed of NGS allows samples to be analyzed fairly quickly, which means that DNA analysis can
70
be practical in a medical setting. By doing this, a doctor can receive the fully sequenced DNA of
71
a patient, and with proper annotation of genetic variants, might be able to use these SNPs to
72
diagnose or guide treatment for a specific disease.
73
Genomic sequencing is already proving useful in the diagnosis and management of allergic and
74
immunologic diseases. In a meta-analysis of studies performing NGS to diagnose primary
75
immunodeficiencies, they found that the proportion of patients with primary
76
immunodeficiencies that were genetically diagnosed ranged from 15-79% of the studies. Early
77
evidence is gathering for genetic biomarkers of allergic disease, including asthma,10 food
78
allergy,11 atopic dermatitis,12 allergic rhinitis,13 and drug allergy.13, 14 However, despite the
79
association of these genetic variants with allergic diseases, the impact on how the disease
80
manifests or varies is often unknown, and the effect of the individual variant is often small. As
81
a result, using genomic data by itself can often only provide an incomplete picture of the
82
individual patient’s personalized risk of disease.15
83
Transcriptomics
84
Transcriptomics involves the characterization of primarily messenger RNA (mRNA) transcript
85
expression levels, most commonly using techniques such as microarray chips or RNA-seq.1
86
Unlike DNA sequencing, transcriptomics provides a potentially more relevant glimpse of the
87
genes actively being transcribed to eventually form proteins. Until recently, early
88
transcriptomics involved the use of oligonucleotide microarrays, chips that contained a set of
5 89
relevant complementary DNA (cDNA) probes. RNA samples were added to bind or hybridize to
90
the matching cDNA probes, and the amount of bound RNA was measured to determine RNA
91
expression levels. While affordable, these techniques were limited by pre-selected cDNA
92
probes, and measurement of new or rare transcripts was not possible. Current advances in NGS
93
technology allows for RNA-sequencing (RNA-seq), where RNA is digested, rapidly sequenced,
94
and then matched up against reference genomes. While this alignment to identify RNA
95
sequences is more computationally intense, it allows better resolution of rare and unknown
96
RNA transcripts.16
97
In the field of allergy and immunology, early research is indicating that transcriptomic changes
98
can be associated with allergic disease phenotype.16-19 For example, one study examining
99
patients with asthma being treated with anti-IL13 therapies described the use of
100
transcriptomics to identify genes whose expression levels predicted improved treatment
101
response, and may potentially be used as a biomarker of therapeutic response.18 Another study
102
examined esophageal biopsies in patients with newly diagnosed eosinophilic esophagitis, and
103
found differential expression of genes in association with esophageal narrowing and with
104
fibrostenosis. These findings may guide the development of targeted treatment modalities and
105
may distinguish which patients are more likely to respond to such therapies.19
106
Epigenomics
107
Epigenomics is defined as the quantification and collective study of the modifications made to
108
DNA and histones, such as methyl tags used by the body to inhibit certain genes.1 While
109
mutations in DNA are the primary means by which individual differences are encoded, these
110
modifications to DNA can also be inherited. Types of DNA modification include the addition of
6 111
methyl or hydroxymethyl groups to the DNA which generally decrease expression of a gene
112
(DNA methylation or hydroxymethylation); the addition of acetyl, methyl, phosphorus or
113
ubiquitin tails to histones to alter transcription (histone modification), and the binding of small
114
noncoding RNAs, known as microRNAs (miRNA), that bind to and block translation of mRNA.16
115
Research in epigenomics has identified novel and heritable changes in DNA methylation,
116
including associations with maternal smoking and air pollution in asthma,20, 21 oral
117
immunotherapy in the development of tolerance in peanut allergy,22, as well as the
118
development of natural tolerance in cow’s milk allergy.23 Epigenetic changes have also been
119
identified in a variety of other allergic diseases including atopic dermatitis,24 allergic rhinitis,25
120
and aspirin-exacerbated respiratory disease.26
121
Proteomics
122
Proteomics is the global characterization of proteins and peptides that are either produced or
123
modified by an organism.1 As proteins are the key product of DNA and RNA and mediate most
124
cellular processes, it is reasonable to expect that proteomics might provide the most accurate
125
reflection of disease pathophysiology.27 However, a number of factors limit the feasibility of
126
high throughput efficient proteomic sequencing, including the complexity of protein structure,
127
the importance of 3-D conformation in mediating protein function, and the large range in
128
concentration. Mass spectrometry, chromatography, and gel electrophoresis are the most
129
common analysis tools at the moment for these set of chemicals, but these techniques can miss
130
key aspects of protein folding. Emerging research have used proteomics techniques to identify
131
subtypes of disease and also examine allergen protein structures. Proteomics approaches have
132
yielded new insights into a variety of allergens, including the allergenicity of milk protein in
7 133
baked milk products,28 identification of B-cell epitopes on fish allergen,29 novel allergens in
134
shrimp,30 as well as simulations of the effects of gastric digestion on peanut component
135
proteins.31 Several studies have begun to use proteomics to analyze subtypes of disease,
136
examining nasal secretions from patients with chronic rhinosinusitis,32, 33 as well as sputum
137
samples from subjects with asthma.34 Overall though, in relation to allergies and personalized
138
medicine, the field of proteomics is still developing and will likely require further advances in
139
proteomic sequencing technology.
140
Metabolomics
141
Metabolomics is the comprehensive study of all the small molecules, known as metabolites,
142
found within a biological specimen that include amino acids, fatty acids, carbohydrates, or
143
other products of cellular metabolic functions. Current technologies now allow a feasible, high-
144
throughput assessment of a large number metabolites resulting from genomic, transcriptomic,
145
and proteomic variability. Metabolomics provides the most integrated profile of biological
146
status reflecting the ‘net results’ of genetic and environmental interactions. 35-37 Compared with
147
other omics, that translation of quantified metabolites into the clinical setting is often very
148
straightforward, as measures of several metabolites are already utilized to diagnose (e.g. blood
149
glucose levels for diabetes) and assess the severity of (e.g. measures of cholesterol) disease.38
150
As technologies continue to expand the scope of measurable metabolites, the potential for
151
metabolites in personalized medicine continues to expand. While the research to identify
152
allergy-associated metabolites is just beginning, metabolites in urine, plasma and stool have all
153
been associated with allergic phenotypes, including intestinal sphingolipids, plasma and dietary
154
polyunsaturated fatty acids, and folate metabolites.39-42 Utilizing multiple metabolomes to
8 155
study allergic disease, particularly food allergies, represents a particular area for potential
156
breakthrough discoveries by providing biological insight into the process of food digestion
157
through the stool metabolome and the subsequent systemic effects in the circulation through
158
plasma/serum metabolomes.
159
Microbiome
160
Microbiomics encompasses the characterization of bacteria and microorganisms living on an
161
organism, and can include flora from a variety of sources including the gut, skin, and mucosal
162
surfaces.1 Early microbiome work relied on a methodology known as 16S sequencing, where
163
ribosomal RNA units from the bacteria were amplified and sequenced.1 These data were then
164
used to cluster species into groups known as OTUs, operational taxonomic units. More
165
recently, a technique known as shotgun sequencing has been developed, where all of the
166
microbial DNA is sequenced, allowing for greater distinction between species.1 Microbiome
167
research has explored dysbiosis, pathologic alterations in gut microbiota, in a number of allergic
168
diseases, including asthma and food allergies. Some genera of bacteria have been increased in
169
association with asthma, including Haemophilus, Moraxella, Neisseria, and Streptococcus.43
170
Furthermore, certain microbial signatures have been linked with specific features of asthma
171
pathophysiology, such as corticosteroid responsiveness and obesity-related asthma.43 In food
172
allergy, dysbiosis resulting in the loss of certain Clostridial species is thought to mediate a loss
173
of regulatory T cell activity that normally promotes the development of tolerance to allergens.44
174
These findings support the potential of microbiome based therapeutic strategies for food
175
allergies.
176
Exposome
9 177
The exposome represents the entirety of exposures that can influence an organism’s
178
development and pathophysiology.45 This term not only encompasses environmental
179
exposures, but can also include aspects like diet and behavioral risk factors that influence
180
disease.46 From an allergist’s perspective, it is fairly intuitive that exposures can influence
181
disease pathophysiology, whether it is the influence of environmental pollen in triggering
182
allergic rhinitis47 or the behavioral and diet effects of early peanut consumption in reducing risk
183
of peanut allergy.48 The field of exposomics is complicated by the wide variety of exposures
184
(biological, chemical, physical, or behavioral) and sources (air, diet, medicinal, soil, water, or
185
even commercial products we use).49 Furthermore, age, developmental stage, and route by
186
which these exposures are encountered can have dramatic effects on disease
187
pathophysiology,49 as seen with food allergy where cutaneous exposure can cause allergic
188
sensitization while oral exposure results in tolerance.50, 51 Measurement of the exposome can
189
encompass a wide variety of techniques, including air/indoor pollutants, allergen sampling,
190
examination of house dust, exposure to food contaminants, and many others.46 Despite these
191
complexities, the Practall consensus group from both the American Academy of Allergy,
192
Asthma, and Immunology and the European Academy of Allergy and Clinical Immunology have
193
identified a need for large-scale initiatives to allow for better characterization of the exposome
194
in the field of allergy.49
195
A number of studies have been performed on the influence of the exposome on allergic
196
disease, though literature searches of the term “exposome” are complicated by the fact that
197
many of these studies were published prior to the coining of the term. Regarding atopic
198
dermatitis, exposure to outdoor air pollutants, volatile organic compounds, and tobacco smoke
10 199
has been associated with worsening atopic dermatitis.52 The effects of pollutants, components
200
of tobacco smoke, bisphenol A, and phthalate metabolites, have also been studied in relation to
201
worsening lung function and asthma.49 Exposure to short chain fatty acids produced in the gut
202
can epigenetically modify DNA and are a mechanism through which diseases like asthma and
203
allergies can form.53 While complicated to measure and characterize, the exposome interacts
204
closely with the –omes described above, and is an essential part of allergic disease
205
development.
206
Integrative Omics
207
Although important insights into allergy and immunology have been gained by the study of
208
single omic data types, it is increasingly recognized the integration of multiple hierarchical
209
levels of omic data has potential to provide the most complete biological insight into allergy
210
pathobiology. Allergic disease affects whole body physiology, via inflammation, oxidative stress
211
and tissue remodeling, and as such is evident and measurable at all levels of the central dogma
212
of molecular biology and now extending well beyond, including the genome, the epigenome,
213
the transcriptome, the metabolome, the exposome, and the micobiome.54 Integrative omic
214
approaches are now utilized to evaluate multiple forms of omic data together and more
215
recently have become a focus in allergy-based research.42, 54, 55 Statistical methods to integrate
216
multiomic data are emerging and multiple review articles have summarized these approaches1,
217
56
218
in epidemiological studies. In the field of allergy and immunology multiomic integration is
219
starting to provide important insights into disease pathophysiology.37 Using both microbiome
220
and metabolomics data, a recent study identified that intestinal microbial-derived sphingolipids
and provide additional insight into important considerations when analyzing multiomic data
11 221
are inversely associated with food allergy.42 Other studies have integrated genetic and
222
metabolomic data using networks and identified important biological relationships between a
223
well-established asthma gene, ORMDL3, lipid metabolism, and asthma phenotypes.
224
It is widely accepted that allergic disease is highly heterogeneous in nature and results from
225
the complex interplay of multiple multiomic influences. Identifying these allergic “endotypes”,
226
i.e. subtypes defined by their functional or pathobiological mechanisms, is crucial as individuals
227
with allergy may appear phenotypically similar but have different underlying pathobiology and
228
respond differently to the same therapy.12 Treatment and management strategies based on
229
underlying pathobiological mechanism, rather than a ‘one-size-fits-all’ approach, may be more
230
effective in terms of improved outcomes and optimized use of healthcare resources. Omics
231
data provide a novel opportunity for endotyping that moves beyond clinical characteristics such
232
as eosinophil count, cytokine count, and IgE levels, particularly when combined with machine
233
learning clustering approaches.57 Utilizing multiomic data in conjunction with clustering
234
algorithms enables the identification of more homogeneous allergy endotypes that are more
235
likely to respond more similarly to specific more personalized treatment regimen. The
236
identification of allergy endotypes utilizing multiomic data, in conjunction with clinical and
237
demographic information, offers one of the most promising approaches creating effective
238
personalized treatments based on molecular data (Figure 3). While molecular endotypes have
239
been created using singular omics data, multiomic molecular endotypes is an emerging field.37,
240
58
241
the molecular endotypes in multiple, diverse populations and repeatedly demonstrate the
242
efficacy of endotype-specific treatment responses.
In order for allergy endotypes to be successfully utilized in the clinic, it is essential to validate
12 243
Limitations of Omics Research
244
As beneficial as omics data may be to personalized medicine, there are still important
245
limitations of these data. It is important to recognize that the precise generation of multiomic
246
data is rapidly evolving. While some fields, such as genetics, have entered an era where whole
247
genome sequencing is a reality, technologies to generate other omic datatypes are more
248
complex and continue to evolve. Other omics, such as the exposome, are inherently difficult to
249
define and measure comprehensively. The multiomic era also comes with the tremendous
250
challenge of “Big Data,” that requires sophisticated analytical techniques and high computing
251
power to identify the disease associated variants. The large amount of data increases the risk of
252
false positive findings, which then necessitates a significantly larger sample size. In rare
253
diseases, it can be difficult to develop sample sizes large enough to overcome these statistical
254
issues. Along with the large sample size, the technical expertise required to analyze these
255
complex data can also add to the cost of conducting robust multi-omics studies. Furthermore,
256
most cohorts have a limited amount of multiomic data; therefore, comprehensive studies that
257
utilize a diverse number of multiomic datatypes together rarely exist. As multiomic data
258
continue to be generated, our ability to identify and replicate allergy associated variants will
259
continue to improve. Simultaneously, statistical methods will continue to evolve and enable
260
more effective analyses with these data, leading to reliable personalized approaches to
261
treatment.
262
Conclusion
263
Multiomic data have great potential to improve our treatment of allergic disease though the
264
implementation of personalized medicine approaches that account for an individual’s
13 265
underlying biological makeup and history of environmental exposures. Continued scientific
266
research in epidemiological studies and clinical trials will likely identify and validate important
267
multiomic variants important for allergic disease and allergy endotypes that have important
268
implications on treatment. Following these discoveries, a critical step for the successful
269
implementation into the clinic is the ability to simply and inexpensively measure important
270
molecular variants for allergy among the patient population. As multiomic knowledge for
271
allergic disease continues to expand, the potential for personalized approach to allergy will
272
likely become a reality.
1
References 1. 2. 3. 4. 5.
6. 7. 8. 9. 10. 11. 12.
13.
14.
15. 16. 17.
18. 19. 20.
21.
Hasin Y, Seldin M, Lusis A. Multi-omics approaches to disease. Genome biology. 2017;18:83. Cobb M. 60 years ago, Francis Crick changed the logic of biology. PLoS Biol. 2017;15:e2003243. McCarthy BJ, Holland JJ. Denatured DNA as a direct template for in vitro protein synthesis. Proc Natl Acad Sci U S A. 1965;54:880-886. Breitling R. What is systems biology? Front Physiol. 2010;1:9. Yska HAF, Elsink K, Kuijpers TW, Frederix GWJ, van Gijn ME, van Montfrans JM. Diagnostic Yield of Next Generation Sequencing in Genetically Undiagnosed Patients with Primary Immunodeficiencies: a Systematic Review. J Clin Immunol. 2019. Mathur S, Sutton J. Personalized medicine could transform healthcare. Biomed Rep. 2017;7:3-5. Bernstein IL, Li JT, Bernstein DI, et al. Allergy diagnostic testing: an updated practice parameter. Ann Allergy Asthma Immunol. 2008;100:S1-148. Shendure J, Findlay GM, Snyder MW. Genomic Medicine-Progress, Pitfalls, and Promise. Cell. 2019;177:45-57. Genomes Project C, Auton A, Brooks LD, et al. A global reference for human genetic variation. Nature. 2015;526:68-74. Davis JS, Weiss ST, Tantisira KG. Asthma Pharmacogenomics: 2015 Update. Curr Allergy Asthma Rep. 2015;15:42. Suaini NHA, Wang Y, Soriano VX, et al. Genetic determinants of paediatric food allergy: A systematic review. Allergy. 2019. Ghosh D, Bernstein JA, Khurana Hershey GK, Rothenberg ME, Mersha TB. Leveraging Multilayered "Omics" Data for Atopic Dermatitis: A Road Map to Precision Medicine. Front Immunol. 2018;9:2727. Waage J, Standl M, Curtin JA, et al. Genome-wide association and HLA fine-mapping studies identify risk loci and genetic pathways underlying allergic rhinitis. Nature genetics. 2018;50:1072-1080. Amo G, Marti M, Garcia-Menaya JM, et al. Identification of Novel Biomarkers for Drug Hypersensitivity After Sequencing of the Promoter Area in 16 Genes of the Vitamin D Pathway and the High-Affinity IgE Receptor. Front Genet. 2019;10:582. Shastry BS. SNPs: impact on gene function and phenotype. Methods Mol Biol. 2009;578:3-22. Bunyavanich S, Schadt EE. Systems biology of asthma and allergic diseases: a multiscale approach. J Allergy Clin Immunol. 2015;135:31-42. Virkud YV, Kelly RS, Croteau-Chonka DC, et al. Novel eosinophilic gene expression networks associated with IgE in two distinct asthma populations. Clin Exp Allergy. 2018;48:1654-1664. Choy DF, Jia G, Abbas AR, et al. Peripheral blood gene expression predicts clinical benefit from anti-IL-13 in asthma. J Allergy Clin Immunol. 2016;138:1230-1233 e1238. Dellon ES, Selitsky SR, Genta RM, Lash RH, Parker JS. Gene expression-phenotype associations in adults with eosinophilic esophagitis. Dig Liver Dis. 2018;50:804-811. Gehring U, Wijga AH, Hoek G, et al. Exposure to air pollution and development of asthma and rhinoconjunctivitis throughout childhood and adolescence: a populationbased birth cohort study. Lancet Respir Med. 2015;3:933-942. Gruzieva O, Xu CJ, Breton CV, et al. Epigenome-Wide Meta-Analysis of Methylation in Children Related to Prenatal NO2 Air Pollution Exposure. Environ Health Perspect. 2017;125:104-110.
2 22.
23. 24. 25.
26. 27.
28. 29.
30.
31.
32.
33.
34. 35. 36.
37. 38. 39. 40.
41.
Syed A, Garcia MA, Lyu SC, et al. Peanut oral immunotherapy results in increased antigen-induced regulatory T-cell function and hypomethylation of forkhead box protein 3 (FOXP3). J Allergy Clin Immunol. 2014;133:500-510. Paparo L, Nocerino R, Cosenza L, et al. Epigenetic features of FoxP3 in children with cow's milk allergy. Clin Epigenetics. 2016;8:86. Thurmann L, Grutzmann K, Klos M, et al. Early-onset childhood atopic dermatitis is related to NLRP2 repression. J Allergy Clin Immunol. 2018;141:1482-1485 e1416. North ML, Jones MJ, MacIsaac JL, et al. Blood and nasal epigenetics correlate with allergic rhinitis symptom development in the environmental exposure unit. Allergy. 2018;73:196-205. Dahlin A, Weiss ST. Genetic and Epigenetic Components of Aspirin-Exacerbated Respiratory Disease. Immunol Allergy Clin North Am. 2016;36:765-789. Betzen C, Alhamdani MS, Lueong S, Schroder C, Stang A, Hoheisel JD. Clinical proteomics: promises, challenges and limitations of affinity arrays. Proteomics Clin Appl. 2015;9:342-347. Bavaro SL, De Angelis E, Barni S, et al. Modulation of Milk Allergenicity by Baking Milk in Foods: A Proteomic Investigation. Nutrients. 2019;11. Carrera M, Gonzalez-Fernandez A, Magadan S, et al. Molecular characterization of Bcell epitopes for the major fish allergen, parvalbumin, by shotgun proteomics, proteinbased bioinformatics and IgE-reactive approaches. J Proteomics. 2019;200:123-133. Lee CH, Wu CC, Tyan YC, Yu WT, Huang ES, Yu HS. Identification of pyruvate kinase as a novel allergen in whiteleg shrimp (Litopenaeus vannamei) by specific-IgE present in patients with shrimp allergy. Food Chem. 2018;258:359-365. Prodic I, Stanic-Vucinic D, Apostolovic D, et al. Influence of peanut matrix on stability of allergens in gastric-simulated digesta: 2S albumins are main contributors to the IgE reactivity of short digestion-resistant peptides. Clin Exp Allergy. 2018;48:731-740. Kim YS, Han D, Kim J, et al. In-Depth, Proteomic Analysis of Nasal Secretions from Patients With Chronic Rhinosinusitis and Nasal Polyps. Allergy Asthma Immunol Res. 2019;11:691-708. Yan B, Lou H, Wang Y, et al. Epithelium-derived cystatin SN enhances eosinophil activation and infiltration through IL-5 in patients with chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol. 2019. Schofield JPR, Burg D, Nicholas B, et al. Stratification of asthma phenotypes by airway proteomic signatures. J Allergy Clin Immunol. 2019;144:70-82. Nicholson JK, Wilson ID. Opinion: understanding 'global' systems biology: metabonomics and the continuum of metabolism. Nat Rev Drug Discov. 2003;2:668-676. Raamsdonk LM, Teusink B, Broadhurst D, et al. A functional genomics strategy that uses metabolome data to reveal the phenotype of silent mutations. Nat Biotechnol. 2001;19:45-50. Reinke SN, Gallart-Ayala H, Gomez C, et al. Metabolomics analysis identifies different metabotypes of asthma severity. Eur Respir J. 2017;49. Clarke SF, Foster JR. A history of blood glucose meters and their role in self-monitoring of diabetes mellitus. Br J Biomed Sci. 2012;69:83-93. McGowan EC, Hong X, Selhub J, et al. Association Between Folate Metabolites and the Development of Food Allergy in Children. J Allergy Clin Immunol Pract. 2019. Inagaki S, Maeda S, Narita M, et al. Urinary PGDM, a prostaglandin D2 metabolite, is a novel biomarker for objectively detecting allergic reactions of food allergy. J Allergy Clin Immunol. 2018;142:1634-1636 e1610. Lee-Sarwar K, Kelly RS, Lasky-Su J, et al. Dietary and Plasma Polyunsaturated Fatty Acids Are Inversely Associated with Asthma and Atopy in Early Childhood. J Allergy Clin Immunol Pract. 2019;7:529-538 e528.
3 42.
43.
44. 45.
46. 47.
48. 49.
50.
51. 52. 53. 54. 55. 56. 57. 58.
Lee-Sarwar K, Kelly RS, Lasky-Su J, et al. Intestinal microbial-derived sphingolipids are inversely associated with childhood food allergy. J Allergy Clin Immunol. 2018;142:335338 e339. Huang YJ, Marsland BJ, Bunyavanich S, et al. The microbiome in allergic disease: Current understanding and future opportunities-2017 PRACTALL document of the American Academy of Allergy, Asthma & Immunology and the European Academy of Allergy and Clinical Immunology. J Allergy Clin Immunol. 2017;139:1099-1110. Stephen-Victor E, Chatila TA. Regulation of oral immune tolerance by the microbiome in food allergy. Curr Opin Immunol. 2019;60:141-147. Wild CP. Complementing the genome with an "exposome": the outstanding challenge of environmental exposure measurement in molecular epidemiology. Cancer Epidemiol Biomarkers Prev. 2005;14:1847-1850. Miller GW, Jones DP. The nature of nurture: refining the definition of the exposome. Toxicol Sci. 2014;137:1-2. Joint Task Force on Practice P. Allergen immunotherapy: a practice parameter. American Academy of Allergy, Asthma and Immunology. American College of Allergy, Asthma and Immunology. Ann Allergy Asthma Immunol. 2003;90:1-40. Du Toit G, Roberts G, Sayre PH, et al. Randomized trial of peanut consumption in infants at risk for peanut allergy. N Engl J Med. 2015;372:803-813. Agache I, Miller R, Gern JE, et al. Emerging concepts and challenges in implementing the exposome paradigm in allergic diseases and asthma: a Practall document. Allergy. 2019;74:449-463. Strid J, Hourihane J, Kimber I, Callard R, Strobel S. Epicutaneous exposure to peanut protein prevents oral tolerance and enhances allergic sensitization. Clin Exp Allergy. 2005;35:757-766. Sicherer SH, Sampson HA. Food allergy: recent advances in pathophysiology and treatment. Annu Rev Med. 2009;60:261-277. Stefanovic N, Flohr C, Irvine AD. The Exposome in Atopic Dermatitis. Allergy. 2019. McKenzie C, Tan J, Macia L, Mackay CR. The nutrition-gut microbiome-physiology axis and allergic diseases. Immunol Rev. 2017;278:277-295. Kelly RS, Dahlin A, McGeachie MJ, et al. Asthma Metabolomics and the Potential for Integrative Omics in Research and the Clinic. Chest. 2017;151:262-277. Kelly RS, Chawes BL, Blighe K, et al. An Integrative Transcriptomic and Metabolomic Study of Lung Function in Children With Asthma. Chest. 2018;154:335-348. Chu SH, Huang M, Kelly RS, et al. Integration of Metabolomic and Other Omics Data in Population-Based Study Designs: An Epidemiological Perspective. Metabolites. 2019;9. Tyler SR, Bunyavanich S. Leveraging -omics for asthma endotyping. J Allergy Clin Immunol. 2019;144:13-23. Kaur R, Chupp G. Phenotypes and endotypes of adult asthma: Moving toward precision medicine. J Allergy Clin Immunol. 2019;144:1-12.
Figure 1: Central Dogma of Molecular Biology. The “central dogma” of molecular biology illustrates the process of changing DNA to RNA to proteins to metabolites via transcription and translation. Figure 1 was created with Biorender.com.
Figure 2: Omes and Omics. Various types of omic data are commonly used in human medical research. The areas of omic research include the omic data generated via the central dogma: genomics (DNA), transcriptomics (RNA), proteomics (proteins), and metabolomics (small molecules including Amino Acids, Fatty Acids, Carbohydrates, Vitamins, Lipids, Nucleotides); however, new types of omic data have emerged, including the fields of epigenomics (methyl tags and histones), exposomics (allergens, toxins, diet, behavior, pollution, climate, radiation, medication), and microbiomics (bacteria and microorganisms). Figure 2 was created with Biorender.com. Figure 3: Omics and Personalized Medicine. As cost of high throughput sequencing decreases and computational methods to analyze these big datasets improve, we may be able to combine various omics datasets to analyze individuals. These omics data can analyzed for biomarkers (a single gene, transcript, epigenetic modification, protein, metabolite, bacteria, or exposure that is associated with disease), or signatures (a combination of many biomarkers), or networks (a complex mapping of many signatures accounting for the relationship between biomarkers within the signatures). These types of analyses will be able to guide diagnostic approaches and identify therapeutic targets tailored to the individual needs of each patient, thereby providing personalized medicine for a variety of allergy endotypes, and distinguishing those without allergy who do not require treatment. Figure 3 was created with Biorender.com.
DNA
Transcription (DNA RNA)
RNA
Translation (RNA Proteins)
Proteins
Processing & Degradation (Proteins Metabolites)
Metabolites
Genome
Epigenome
Transcriptome
Proteome
Metabolome
Microbiome
Exposures
Metabolites
DNA (genes)
Methyl Tags and Histones
RNA (transcripts)
Proteins
Genomics
Epigenomics
Transcriptomics
Proteomics
Exposome
(Amino Acids, Fatty Acids, Carbohydrates, Vitamins, Lipids, Nucleotides)
Bacteria and Microorganisms
(Allergens, Toxins, Diet, Behavior, Pollution, Climate, Radiation, Medicines)
Metabolomics
Microbiomics
Exposomics
Patients
Omic Datatypes
Bioinformatics Analysis
Personalized Medicine Allergy Endotypes
Genomics
Biomarker Epigenomics Signature
Transcriptomics
Proteomics Network
Metabolomics
Microbiomics Exposomics
Non-Allergic / No Treatment
Key Messages •
• •
•
The term “omics” (aka multi-omics) refers to the usage of high-throughput technology to characterize a set of molecules relevant to a field of biology, and has grown to encompass a variety of “-omes” including the genome (DNA), transcriptome (RNA), proteome (proteins), metabolome (metabolites), microbiome (microbiota), exposome (exposures) Integrative omics refers to the study of how different omics data interact to affect a condition or disease of interest. Personalized medicine is an approach to studying disease etiology, pathogenesis, progression and treatment, that utilizes information specific to an individual (for example, their genome or transcriptome) to identify the optimal treatment option for that individual. Endotype refers to a subtype of a disease that is defined by a distinct pathophysiologic mechanism, in contrast with a phenotype, which describes characteristics of a disease without implying a different underlying mechanism.
The nuts and bolts of omics for the clinical allergist Authors: Yamini V. Virkud M.D.1, 2 Rachel S. Kelly Ph.D.2, Caleb Wood*, Jessica A. Lasky-Su Sc.D.2 Affiliations: 1. Department of Pediatrics, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, USA 2. Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA * Indicates no academic degree Corresponding author: Jessica Lasky-Su Journal: Annals of Allergy, Asthma, and Immunology Address: 181 Longwood Avenue, Boston, MA 02115 Funding: This manuscript is supported by an R01 grants from the National Heart, Lung, and Blood Institute of the National Institutes (NHLBI) of Health to Brigham and Women’s Hospital, NHLBI 1R01HL123915-01 and R01HL141826. (JALS). YVV. was supported by a grant from the National Institute of Allergy and Infectious Diseases of the US National Institutes of Health (K23AI130408). RSK is supported by the grant from NHLBI (K01HL146980). Keywords: Allergy, personalized medicine, precision medicine, omics, microbiome, metabolomics, transcriptomics, genomics, exposome, computational biology, systems biology, endotype; gene expression Abbreviations: Clinical Trial or Protocol number associated with this study: N/A Running Title: Omics for the Clinical Allergist Manuscript Body Word Count: 3,283 Author Contributions: Yamini Virkud - literature review, writing the manuscript, figure generation Rachel S. Kelly - literature review, writing the manuscript, figure generation Caleb Wood - literature review, writing the manuscript, figure generation Jessica A. Lasky-Su - literature review, writing the manuscript, figure generation Conflict of Interest: None of the authors involved in this manuscript have any conflicts of interest relevant to this work