1/15https://immunenetwork.org
ABSTRACT
Th cells, which orchestrate immune responses to various pathogens, dierentiate from
naïve CD4 T cells into several subsets that stimulate and regulate immune responses against
various types of pathogens, as well as a variety of immune-related diseases. Decades of
research have revealed that the fate decision processes are controlled by cytokines, cytokine
receptor signaling, and master transcription factors that drive the dierentiation programs.
Since the Th1 and Th2 paradigm was proposed, many subsets have been added to the list.
In this review, I will summarize these events, including the fate decision processes, subset
functions, transcriptional regulation, metabolic regulation, and plasticity and heterogeneity.
I will also introduce current topics of interest.
Keywords: Helper T cell; T-lymphocyte subset; Cell dierentiation; Adaptive immune response;
Transcription factors; Cytokines
INTRODUCTION
Pathogens can infect many dierent tissues and cellular compartments, and the life cycles
of these pathogens can vary considerably. To overcome this, the host immune system
has developed pathogen-specic strategies. For example, Th cells (eector CD4 T cells)
orchestrate immune responses against many dierent pathogens (1-4). Naïve CD4 T cells
recirculate via the blood and lymphatic systems, gathering information about pathogens by
binding to antigens presented by dendritic cells (DCs) in lymphoid tissues. DCs also secrete
cytokines when they present these pathogen-specic antigens to naïve CD4 T cells, thereby
triggering dierentiation pathways (1-4). Activated CD4 T cells proliferate and dierentiate
into dierent immune cell subsets, which determine the type of immune response (1-4).
Research in the eld of Th cell dierentiation and function began about 30 years ago, and it
is still progressing. In this review, I will summarize the basic principles that govern Th cell
dierentiation (Fig. 1). Other topics in this eld, including the microbiota and plasticity/
heterogeneity, will be covered by other articles in this issue.
Immune Netw. 2023 Feb;23(1):e4
https://doi.org/10.4110/in.2023.23.e4
pISSN 1598-2629·eISSN 2092-6685
Review Article
Molecular Mechanisms of T Helper
Cell Differentiation and Functional
Specialization
Received: Nov 30, 2022
Revised: Jan 17, 2023
Accepted: Jan 29, 2023
Published online: Feb 17, 2023
*Correspondence to
Gap Ryol Lee
Department of Life Science, Sogang University,
35 Baekbeom-ro, Mapo-gu, Seoul 04107,
Korea.
Copyright © 2023. The Korean Association of
Immunologists
This is an Open Access article distributed
under the terms of the Creative Commons
Attribution Non-Commercial License (https://
creativecommons.org/licenses/by-nc/4.0/)
which permits unrestricted non-commercial
use, distribution, and reproduction in any
medium, provided the original work is properly
cited.
ORCID iDs
Gap Ryol Lee
https://orcid.org/0000-0002-6412-9482
Conflict of Interest
The authors declare no potential conflicts of
interest.
Abbreviations
AMPK, AMP-activated protein kinase; APC,
antigen-presenting cell; DC, dendritic
cell; EAE, experimental autoimmune
encephalomyelitis; LCR, locus control
region; OXPHOS, oxidative phosphorylation;
Phd, prolyl-4-hydroxylase domain; pTreg,
regulatory T cell in the periphery; scRNA-seq,
single cell RNA sequencing; T, follicular
helper T; Tr1, type-1 regulatory T; tTreg,
regulatory T cell development in the thymus;
Vhl, von Hippel-Lindau.
Gap Ryol Lee
*
Department of Life Science, Sogang University, Seoul 04107, Korea
BRIEF HISTORY OF Th SUBSETS
For a long time, Th cells were regarded as cells that aid activation of B cells and macrophages
by producing cytokines. In 1986, Mosmann and colleagues (5) published a seminal work
showing that Th cells comprise 2 distinct cell types based on their cytokine proles, namely,
Th1 and Th2. The dierent cytokines produced by these cell types lead to mutually exclusive
functional properties (5). Th1 cells activate macrophages and mediate immune responses
against intracellular pathogens, while Th2 cells stimulate B cells (5). Identication of Th
cell heterogeneity and dierences in functional properties was a major conceptual advance
in the eld, and triggered further study of the molecular mechanisms that regulate the
fate-determining processes and functional properties. These eorts led to identication of
cytokines, signaling pathways, and transcription factors as determinants of Th1 and Th2
subset dierentiation. In 2005, Th17 cells were identied and proposed as an independent
subset (6-8). Th17 cells induce inammation and mediate immune responses against
extracellular bacteria and fungi (9,10). Another independent subset, called follicular helper
T (T) cells, was also identied (11-13). T cells reside in the germinal centers of lymphoid
tissue and interact with B cells to trigger maturation. It was thought that B cell stimulation
was the role of Th2 cells; however, this is now thought to be the role of T cells. In addition
to activator cells, repressor cell types have also been identied; these are termed Treg cells
(14-16). Many other subsets have also been identied, including IL-9-producing Th9 cells, IL-
22-producing Th22 cells, and IL-10 producing type-1 regulatory T (Tr1) cells. Th9, Th22, and
Tr1 cells will not be covered in this review because our understanding of their functions and
roles is still in its infancy.
T Helper Cell Differentiation and Function
https://doi.org/10.4110/in.2023.23.e4
2/15
https://immunenetwork.org
cytokine
receptor
CD28
TCR
nutrients
metabolism
Th1: IL-12, IFN-γ
Th2: IL-4
Th17: TGF-β + IL-6 (IL-21)
Treg: TGF-β
inducer
cytokines
STAT
or SMAD
master
TF
Master TF
Activation/proliferation
differentiation
effctor
cytokine
Th1: STAT4, STAT1
Th2: STAT6
Th17: STAT3
Tfh: STAT3
Treg: SMAD2, SMAD3, STAT5
Th1: T-bet
Th2: GATA3
Th17: RORγt
Tfh: Bcl6
Treg: Foxp3
Th1: IFN-γ
Th2: IL-4, IL-5, IL-13
Th17: IL-17A, IL-17F, IL-22
Tfh: IL-21
Treg: IL-10
differentiation
Figure 1. Schematic diagram of Th cell differentiation. Th cell differentiation pathway is determined by inducer
cytokines that are present during T cell activation. The cytokine signaling activates STAT or SMAD, which in
turn induces master transcription factors. Master transcription factors drive the subset-specific differentiation
programs. Th cell proliferation and differentiation are also influenced by nutrient or metabolites.
Th CELL FUNCTIONS
Eector CD4 T cells either stimulate or repress other immune cells. The most important
mediators of these functions are cytokines (1-4). Th1 cells produce IFN-γ, which stimulates
macrophages and CTLs. Stimulated macrophages kill intracellular pathogens by activating
microbicidal mechanisms, including facilitation of fusion of phagosomes with lysosomes,
activation of NADPH oxidase, and generation of reactive oxygen species (17). Activation of CTL
requires the help of Th1 cells via secretion of IFN-γ and CD40L signaling, which trigger CTL
dierentiation. Th2 cells produce IL-4, IL-5, and IL-13, which mobilize or stimulate eosinophils,
basophils, and mast cells to combat helminthic parasites (18). IL-13 also stimulates intestinal
epithelial cells to increase cell turnover and movement, as well as acting on smooth muscle cells
in the intestine to increase contractility, all of which help to remove parasites from the gut. Th17
cells produce IL-17A, IL-17F, and IL-22, which stimulate epithelial cells, broblasts, and stromal
cells to produce GM-CSF, recruit neutrophils, and induce inammation, thereby mediating
immune responses against extracellular bacteria and fungi (9,10). T cells produce IL-21
and activate B cells in the germinal center to induce maturation via somatic hypermutation
and class-switch recombination (19). Treg cells inhibit eector T cell and B cell responses, as
well as innate immune responses (20). Treg cells produce IL-10, TGF-β, and IL-35 to inhibit
immune responses, although they can use other mechanisms too. Treg cells express high
amounts of IL2Rα on their surface, leading to deprivation of IL-2, which is required for growth
of eector CD4 T cells. Treg cells express coinhibitory receptors such as Ctla-4 and Lag3,
which interfere with the interaction with costimulatory molecules and MHC class II molecules
expressed by antigen-presenting cells (APCs), thereby preventing T cell activation (21,22).
Treg cells also kill CD4 T cells directly by secreting cytotoxins. They also express high amounts
of ectonucleotidases such as CD39 and CD73, leading to production of immunosuppressive
adenosine, which promotes T cell anergy (23). Although Treg development in the thymus
(tTreg) and Treg in the periphery (pTreg) cells have overlapping functions, tTregs are more
specialized at preventing autoimmune responses against self-antigens, whereas pTregs are
more specialized in inducing tolerance to foreign antigens.
Each CD4 T cell subset cross regulates other subsets. For example, Th1 cells inhibit Th2 and
Th17 cell dierentiation by suppressing expression of IL-4 and IL-17, respectively (7,24). Th2
cells repress Th1 dierentiation by inhibiting IL-12Rβ2 and IL-17 expression, respectively
(7,25). T cells inhibit dierentiation of other subsets via Bcl6-mediated repression of
Prdma1
(which encodes Blimp-1) expression (26,27).
In addition to the induction of protective immunity against infectious agents, Th cells are
responsible for the pathology of many other diseases caused by aberrant immune responses
(1-4). For example, Th1 and Th17 cells plays critical roles in rheumatoid arthritis, type I
diabetes, and multiple sclerosis; Th2 cells in allergic diseases such as asthma, rhinitis,
and allergic dermatitis; and Treg cells in autoimmune disease, allergy, and cancer. Thus,
regulating subset-specic functions is an eective strategy for treating these immune-related
diseases. Indeed, based on this knowledge, many therapeutic strategies have been, or are
being, developed (28,29).
T Helper Cell Differentiation and Function
https://doi.org/10.4110/in.2023.23.e4 3/15
https://immunenetwork.org
SIGNAL TRANSDUCTION AND TRANSCRIPTIONAL
REGULATION
Naïve CD4 T cells dierentiate into various subsets of eector cells when they recognize their
cognate antigens presented in conjunction with MHC II molecules on the surface of DCs in
lymphoid tissues (1-3). These subsets include Th1, Th2, Th17, T, and Treg cells (1-3). The
fates of these subsets are determined by the cytokines (produced mainly by APCs) present in
the environment at the time of activation (1-3). The cytokines bind to their cognate receptors
on naïve CD4 T cells and activate the STAT or SMAD pathway (1-3). Th1 cell dierentiation is
induced by IL-12 or IFN-γ, which activate Stat4 or Stat1, respectively (1-3). Th2 cell dierentiation
is induced by IL-4, which activates Stat6 (1-3). Th17 cell dierentiation is induced by TGF-β plus
IL-6 (or IL-21), which activate Stat3 (1-3). T cell dierentiation is induced by IL-21, which also
activates Stat3 (1-3). Treg cells are rather dierent in that they can be generated during T cell
development in the thymus (tTreg cells) or can dierentiate from naïve CD4 pTreg cells (1-3).
In
vitro
, Treg cells can be dierentiated by TGF-β, which activates Smad2 and Smad3 (1-3).
STAT proteins enter the nucleus and induce global epigenetic changes as well as expression
of master transcription factors which drive dierentiation programs (1-4). T-bet drives the
Th1 program, Gata-3 drives the Th2 program, Rorγt drives the Th17 program, Bcl6 drives
the T program, and Foxp3 drives the Treg program. These transcription factors induce
expression of a variety of genes required for dierentiation, and for the eector functions,
of each subset. Division of the roles played by STATs and the master transcription factors is
the focus of much interest. Although both factors are important for dierentiation, it seems
likely that STATs act as pioneering factors that shape global enhancer landscapes and gene
expression patterns, whereas master transcription factors regulate a limited set of genes that
is required for subset-specic dierentiation and function (30,31).
Although master transcription factors drive the dierentiation program in Th cells, many
other transcription factors play important roles in the process (32,33). In addition to the
master transcription factors, Irf1, Runx3, Hlx, Ets-1, Nfatc1, and Bhlhe40 play important roles
in Th1 cell dierentiation; c-Maf, Notch/CSL, IRF4, G-1, and Yy1 in Th2 dierentiation; Ifr4,
Batf, and Runx1 in Th17 dierentiation; and Runx1, Nr4a2, Foxo family members, Satb1, and
Helio in Treg dierentiation (32,33). These transcription factors form a coordinated network
that drives the dierentiation programs (34-36), even in the absence of master transcription
factors (36). The roles of these transcription factors depend on individual factors. In general,
these factors either enhance transcription, induce changes in epigenetic status, or promote
organization of the relevant genes or gene loci within the genome (32,33).
EPIGENETIC REGULATION
Along with transcription factors, the genetic loci of characteristic cytokine genes have been
investigated extensively with respect to their role in regulating Th cell dierentiation (37,38).
This is due in part to the fact that the dierentiation process is easily controlled
in vitro
by
cytokines, enabling investigation of cell fates within a short period of time. Such studies
have reported interesting ndings: 1) epigenetic changes accompany gene expression during
dierentiation; 2) poised state of cytokine loci even before dierentiation; 3) coordinate
expression of cytokine genes; 4) inter-chromosomal interactions between 2 opposing loci;
and 5) insulation of cytokine loci.
T Helper Cell Differentiation and Function
https://doi.org/10.4110/in.2023.23.e4
4/15
https://immunenetwork.org
Epigenetic changes during Th cell differentiation
Marked changes in gene expression occur during CD4 T cell activation and dierentiation.
Many of the genes expressed by activated T cells are required for cell division. Since naïve T
cells are in a quiescent state, transition to actively dividing cells requires systemic changes
in gene expression. Yet more genes are required for subset-specic eector functions; for
example, genes encoding cytokines that mediate the eector functions of a particular subset.
Not surprisingly, naïve CD4 T cells undergo epigenetic changes such as increased accessibility
by DNase I, histone acetylation, and DNA methylation (37,38).
Ifng
and the Th2 cytokine locus
(the
Il4-Il13-Il5
locus) show subset-specic epigenetic changes. During Th1 dierentiation,
the
Ifng
locus opens, but the Th2 cytokine locus remains closed. Likewise, the Th2 cytokine
locus opens during Th2 dierentiation, but the
Ifng
locus remains closed. This subset-specic
chromatin modication is inuenced by master transcription factors such as T-bet and Gata-3.
Thus, one of the roles of these transcription factors is to open the chromatin at the loci of
subset-specic genes to make it “competent” for expression of these genes.
Poised state of cytokine loci before differentiation
Certain genetic loci, such as the
Ifng
locus, are demethylated even before Th1 dierentiation.
This hypomethylation pattern seems to be established early during T cell development,
including that of naïve CD4 T cells, suggesting that these loci are predisposed to expression
(37,38). During Th1 dierentiation, this demethylation pattern at the
Ifng
locus is preserved;
however, the locus becomes methylated during Th2 dierentiation. Genome-wide studies
of the epigenetic status of chromatin and gene expression in CD4 T cells revealed that the
histone modication status of key cytokine and transcription factor loci correlates strongly
with gene expression patterns (39). Interestingly, this study also found that
Tbx21
and
Gata3
loci contain both active (H3K4me3) and suppressive (H3K27me3) histone marks (39). This
bivalent status with respect to histone marks also predispose gene expression, suggesting
that cells are preparing for rapid changes in gene expression that occur during activation and
dierentiation. The poised status of multiple key genetic loci facilitates easy conversion from
one dierentiated state to another.
Coordinate expression of cytokine genes
The
Ifng
locus and the Th2 cytokine locus are regulated by promoters, enhancers, and
silencers in the same manner as other genetic loci (37,38). Since the
Il4
,
Il13
, and
Il5
genes
are clustered, it is of interest to see whether they are coordinately regulated in Th2 cells.
One of the regulatory elements that exerts coordinate expression of clustered genes is the
locus control region (LCR), best exemplied by the β-globin LCR (40). The LCR is dened
as a sequence that enables copy number-dependent expression in transgenic mice (40). The
Th2 cytokine locus contains an LCR, called the Th2 LCR, which is interspersed in the intron
regions of the
Rad50
gene located between the
Il13
and
Il5
genes (41). The Th2 LCR undergoes
epigenetic changes during Th2 dierentiation by recruiting GATA3 and other transcription
factors such as Yy1, Stat6, Satb1, and Irf4 (42-44). This process is essential for Th2 cytokine
expression since deletion of the Th2 LCR or its components abolishes Th2 cytokine expression
(42,45,46). The Th2 LCR induces chromosomal looping to facilitate interactions with the
promoters of Th2 cytokine genes, thereby forming a basis for coordinated regulation (47).
Inter-chromosomal interactions between 2 opposing loci
Chromosomal interactions occur not only intra-chromosomally, but also inter-
chromosomally. Chromosomal interaction between 2 dierent chromosomes has been
suggested for long time, but it was proved experimentally in the study of
Ifng
and Th2 loci,
T Helper Cell Differentiation and Function
https://doi.org/10.4110/in.2023.23.e4 5/15
https://immunenetwork.org
shedding light on a novel mechanism of gene regulation (48). Although they are located
on dierent chromosomes,
Ifng
and RHS6, a component of the Th2 LCR, in naïve CD4 T
cells interact with each other. This physical interaction seems to maintain a poised state
for expression of each gene until the cell is activated, since this interaction is replaced with
intrachromosomal interactions upon Th2 cell dierentiation (48). In naïve CD4 T cells,
RHS6 also interacts with the
Il17
promoter through Oct-1 and CTCF, suggesting occurrence of
inter-chromosomal interactions among multiple chromosomes (49). When Oct-1, CTCF, or
the interacting genetic loci are decient, the interaction does not occur and expression of
Il17
increases (49), supporting its role in cytokine gene regulation.
Insulation of cytokine loci
The
Ifng
locus is located about 40 kb downstream of the
IL22
gene in humans and 245 kb
downstream of the
Il22
gene in mice. Since
Il22
is a Th17-specic gene, expression of
Ifng
and
Il22
should be regulated separately. Indeed, an insulator sequence between these gene loci was
identied by genome-wide screening of CTCF binding sites and cohesion binding sites (50,51).
The
Ifng
locus is insulated by −70 and +66 boundary elements, which contain CTCF and the
cohesion component Rad21 (50,51). Although recruitment of CTCF to the −70 site is conserved
in naïve, Th1, and Th2 cells, CTCF recruitment to the
Ifng
intronic region and +66 site is specic
to Th1 cells (50,51), suggesting that this insulator sequence, and its binding to CTCF, may play a
role in forming the intra-chromosomal loops needed for the
Ifng
gene transcription.
PLASTICITY AND HETEROGENEITY
I will provide a very brief overview of this topic as it is covered in depth by another article in
this issue. Initially, the status of Th1 and Th2 cells was thought to be stable and terminally
dierentiated; this is because reversal between subsets cannot be induced aer long-term
culture (52), and production of specic cytokines seems to be stably maintained (53).
However, later studies showed that Th subsets are not permanently dierentiated; rather
they can dierentiate into another subset under certain circumstances. This phenomenon
is called plasticity (54,55). Early
in vitro
studies suggested that forced expression of master
regulators by already dierentiated cells induces alternative dierentiation pathways (55).
For example, Th1 cells produce IL-4 upon transfection with Gata-3, and Th2 cells can express
IFN-γ upon transfection of T-bet. A fate-mapping study using a mouse model of experimental
autoimmune encephalomyelitis (EAE) revealed that most cells producing IFN-γ previously
expressed IL-17A (56). Studies in animal models of diabetes and cancer show that Th17 cells
acquire Th1-like properties (57,58).
In addition to plasticity, CD4 T cells show functional heterogeneity. Cells co-expressing
T-bet and RORγt are present in the gut and inamed brain tissue (56,59). These cells express
both IFN-γ and IL-17, and are thought to play an important role in immune responses to
Mycobacterium tuberculosis
(60). Treg cells oen co-express T-bet, Gata3, or Rorγt. T-bet-
expressing Treg cells inhibit type I immune responses (61). Gata3 is frequently co-expressed
with Foxp3 by Treg cells and is thought to help control Th2-type immune response (62).
Treg cells co-expressing Irf4 or Stat3 control Th2- and Th17-mediated immune responses,
respectively (63,64). Thus, co-expression of transcription factors by other subsets seems to
endow Treg cells with the ability to control specic eector functions.
T Helper Cell Differentiation and Function
https://doi.org/10.4110/in.2023.23.e4
6/15
https://immunenetwork.org
The plasticity and heterogeneity of Th subsets modies and expands the characteristics
of each subset. Plasticity is thought to be an adaptation to a changing environment or to
pathogens. Since some pathogens change their habitats (e.g., by infecting dierent cells/
organs) and their virulence strategy during their life cycle, the immune system needs to
change its defensive strategies to combat them.
METABOLIC REGULATION
When naïve CD4 T cells are activated and dierentiate into eector cells, they undergo
drastic changes in their metabolic programs (1,65-67). During this process, dierentiating
and proliferating cells require energy, as well as the building blocks to synthesize DNA,
RNA, proteins, and lipids. Eector subsets of Th cells, namely, Th1, Th2, and Th17 cells, rely
more heavily on glycolysis than Treg cells (1,34,65,66). Most cells generate ATP via oxidative
phosphorylation (OXPHOS), which occurs in the mitochondria. However, rapidly dividing
cells such as cancer cells use aerobic glycolysis instead of OXPHOS to generate ATP. Although
it generates less ATP than OXPHOS, aerobic glycolysis can provide many of the materials
required for biosynthesis in cells proliferating under hypoxic conditions. This phenomenon,
called the “Warburg eect,” also occurs in eector T cells, which are rapidly dividing cells. T
cells also show enhanced glycolysis, but cell-autonomous IL-2 inhibits the dierentiation and
metabolic programs of T cells. Th17 cells also use glutaminolysis and fatty acid synthesis
programs, whereas fatty acid synthesis antagonizes dierentiation of Treg cells. By contrast,
Treg cells rely on mitochondrial activity for their dierentiation, homeostasis, and function.
Since Th cell dierentiation requires metabolic reprogramming, factors that regulate this
process aect Th cell dierentiation (65). These factors include 1) nutrients/metabolites, 2)
transcription factors, and 3) signaling molecules.
Nutrients/metabolites
Glucose is a readily available nutrient that provides energy to activated and proliferating T
cells, as well as building blocks for nucleic acids, proteins, lipids, and carbohydrates. Glucose-
6-phosphate, the rst intermediate of glycolysis, can be converted to fructose-6-phosphate
to yield pyruvate, which is used to synthesize glycogen, or shuttled to synthesize purines
and pyrimidines via the pentose phosphate pathway. TCR and/or costimulatory signaling
through CD28 triggers rapid uptake of glucose by upregulating the glucose transporter Glut1
and by increasing glycolytic ux (1,65-67). Thus, activated CD4 T cells are highly dependent
on glucose uptake. Indeed, Th1 and Th17 cells have an increased glycolytic rate. Chemical or
genetic inhibition of specic steps within the glycolytic pathway leads to a marked reduction
in eector T cell proliferation and cytokine production. Deprivation of glucose impairs T cell
activation and survival, highlighting the importance of glucose for the T cell activation and
dierentiation processes (1,34,65,66). Glucose deprivation also inhibits IFN-γ production,
as well as reducing production of phosphoenolpyruvate, which supports Ca
2+
/NFAT signaling
required for glycolytic reprogramming in T cells (1,65-67). Unlike eector T cell subsets,
Treg cells are not aected by Glut1 deciency (1,34,65,66). Blocking glycolysis with 2-DG,
an inhibitor of hexokinase (an enzyme that regulates the rate limiting step of glycolysis),
inhibits dierentiation of CD4 and CD8 T cells into eector cells (1,65-67). Deciency of
lactate dehydrogenase A, an enzyme that converts lactate to pyruvate, inhibits ATP generation
and PI3K/Akt activation, thereby repressing T cell activation (68). High levels of glycolytic
metabolism inhibit induction of Foxp3, instead inducing Th17 cell dierentiation (69).
T Helper Cell Differentiation and Function
https://doi.org/10.4110/in.2023.23.e4 7/15
https://immunenetwork.org
Fatty acid metabolism is a critical part of T cell dierentiation. Fatty acid oxidation is
essential for Treg cell generation and function, and for making membrane components by
proliferating eector T cells, including Th1, Th2, and Th17 cells (70). Proliferating activated
T cells show increased fatty acid synthesis, with a concomitant reduction in fatty acid
oxidation. Fatty acid metabolism is important for the balance between Th17 and Treg cells:
fatty acids aid the generation and function of Treg cells, while controlling the pathogenicity
of Th17 cells (70).
Transcription factors
Myc, induced by TCR stimulation, is essential for metabolic reprogramming of T cell activation
and function. Myc (71), which plays an essential role in glycolysis and glutaminolysis in activated
T cells, induces Glut1 and several glycolytic enzymes (71). When Myc is ablated, glycolysis
is impaired, resulting in decreased T cell activation and growth (72). Myc also stimulates
glutaminolysis by increasing expression of glutaminase and glutamine transporters (73).
HIF-1α plays an important role in Th cell dierentiation and function (74) by enhancing Th17
cell dierentiation via stimulation of
Il17
gene transcription while reducing Treg cell generation
via Foxp3 degradation (75). Conditional deletion of HIF-1α causes resistance to EAE, with
reduced inltration of the CNS by Th17 cells (69). In addition, HIF-1α induces glycolysis under
hypoxic conditions. The oxygen-sensing function is regulated by 2 proteins; the von Hippel-
Lindau (Vhl) complex supports and prolyl-4-hydroxylase domain (Phd) proteins oppose
the function. Upon activation,
Vhl
-decient T cells show augmented glycolysis but reduced
mitochondrial oxidative metabolism, leading to enhanced eector programming, eector/
memory cell dierentiation, and cell death (76,77).
Phd
-decient CD4 T cells, however, show
increased eector cell dierentiation but decreased Treg cell dierentiation (78).
Signaling molecules
The mTOR is a serine/threonine kinase that transmits signals from the PI3K-Akt pathway.
mTOR forms 2 functionally distinct complexes, mTORC1 and mTORC2, and is essential for
regulating glucose metabolism and upregulating glycolysis. It is also required for eector T
cell dierentiation; indeed, mTOR deciency causes failure of eector T cell dierentiation
and leads to Treg cell generation instead (79). mTORC1 and mTORC2 play distinct roles
during dierentiation of Th subsets. CD4 T cells lacking mTORC1 activity due to the absence
of one of its components (Rheb) failed to generate Th1 and Th17 cells (80), whereas those
lacking mTORC2 activity due to the absence of Rictor failed to generate Th2 cells (80,81).
Excessive mTOR activity caused by
Pten
-deciency reduces the stability of Treg cells, leading
to uncontrolled Th1 and T cell responses (82,83).
AMP-activated protein kinase (AMPK) is a glucose-sensitive metabolic sensor that regulates
mRNA translation and glutamine-dependent mitochondrial metabolism (84). AMPK
promotes catabolic processes but inhibits anabolic processes. It also promotes OXPHOS,
mitochondrial biogenesis, fatty acid oxidation, and autophagy, but inhibits glycolysis,
glutaminolysis, and glycogen and fatty acid synthesis (84). AMPK promotes energy
conservation by T cells, as well as maintaining T cell bioenergetics and viability (84). It also
negatively regulates eector Th cell dierentiation by inhibiting mTORC1 activity.
Ampka1
-
decient T cells show reduced metabolic plasticity and mitochondrial bioenergetic when
glucose is limited (84). Under glucose-limiting conditions, reduced expression of IFN-γ
mRNA and protein aects Th1 dierentiation. By contrast, AMPK induces dierentiation of
naïve CD4 T into Treg cells (85).
T Helper Cell Differentiation and Function
https://doi.org/10.4110/in.2023.23.e4
8/15
https://immunenetwork.org
CHALLENGES TO THE Th SUBSET PARADIGM
The paradigm of Th subsets has contributed greatly to our understanding of the immune
system. The paradigm divides immune responses into categories, and provides a simple
framework for understanding otherwise complex phenomena. Based on this paradigm,
many immune-related diseases have been categorized as type I, type II, and type III diseases.
This categorization is also applied to innate immune cells, such as innate lymphoid cells and
macrophages. The paradigm was originally proposed, and mainly studied, in the context of
in
vitro
-dierentiated CD4 T cells, generation of which requires excessive amounts of cytokines
and neutralizing antibodies.
The plasticity and heterogeneity of Th cells modies their functions rather elegantly without
necessarily challenging the classical view of Th cell subsets, which states that these subsets
are terminally dierentiated and stable. Recent technological advances such as single cell
RNA sequencing (scRNA-seq) and CyTOF have made it possible to analyze gene expression
at the single cell level. Studies using these technologies show that eector Th cells are much
more heterogeneous than the paradigm suggests.
scRNA-seq and computational analysis show that Th17 cells isolated from animal models
of autoimmune diseases are heterogeneous and show a spectrum of cellular states (86,87).
Tortola et al. (88) used CyTOF to analyze expression of subset-specic marker proteins
expressed by
in vitro
-dierentiated Th cells and by Th cells in animal disease models. They
found that eector Th cells are far more heterogeneous than expected, even under controlled
stimulation conditions. Kiner et al. (89) analyzed gut CD4 T cells using scRNA-seq and
ATAC-seq. Surprisingly, they found that CD4 T cells do not follow the classic subset-specic
gene expression pattern. IFN-γ and IL-17A are expressed by most eector cells, many of
which co-express both genes (89). Transcriptional and epigenetic patterns are determined
by infectious agents rather than by cytokine and master transcription factor signatures (89).
scRNA-seq followed by trajectory analysis revealed that Th1 cells and T cells bifurcate
during their dierentiation in malaria infection model (90). These results challenge the Th
subset paradigm, and suggest that Th cells exist in a heterogeneous state of dierentiation,
which may change according to the environment. Thus, more studies in this area are needed.
CONCLUSIONS AND PERSPECTIVES
Th cells play essential roles in coordinating immune responses to dierent pathogens.
They are also responsible for many immune pathologies. Thus, research on Th cells (either
the fundamental mechanisms underlying dierentiation and function, or therapeutic
applications based on the results of such studies) is ongoing. New technological advances
will speed up progress. For example, scRNA-seq can reveal detailed gene expression
patterns in individual cells within a large population, providing information about
possible dierentiation pathways. CRISPR/Cas9 technology enables functional study
of genes of interest on a large scale, either
in vitro
or
in vivo
. Introduction of the concept
of super-enhancers, which are dened as regulatory regions that are highly enriched in
transcription factors and activating/suppressive epigenetic marks, will facilitate more rapid
study of regulatory regions, since they can be identied by genome wide approaches and
bioinformatics analysis. These new technologies or concepts will enable comprehensive
analysis of the molecular mechanisms underlying these processes, while challenging the
T Helper Cell Differentiation and Function
https://doi.org/10.4110/in.2023.23.e4 9/15
https://immunenetwork.org
current paradigm. I anticipate and hope that more and more new and interesting ndings
about the underlying molecular mechanisms will come to light, and that we will make
progress in the treatment of dicult-to-cure diseases.
ACKNOWLEDGEMENTS
This work was supported by the National Research Foundation of Korea (NRF) grants
funded by the Korean government (2021K2A9A2A06048161, 2022R1A2B5B03001840,
2022R1A4A5032688 to GRL).
REFERENCES
1. Saravia J, Chapman NM, Chi H. Helper T cell dierentiation.
Cell Mol Immunol
2019;16:634-643.
PUBMED | CROSSREF
2. O’Shea JJ, Paul WE. Mechanisms underlying lineage commitment and plasticity of helper CD4
+
T cells.
Science
2010;327:1098-1102.
PUBMED | CROSSREF
3. Murphy KM, Ouyang W, Farrar JD, Yang J, Ranganath S, Asnagli H, Aarian M, Murphy TL. Signaling and
transcription in T helper development.
Annu Rev Immunol
2000;18:451-494.
PUBMED | CROSSREF
4. Zhu J, Yamane H, Paul WE. Dierentiation of eector CD4 T cell populations (*).
Annu Rev Immunol
2010;28:445-489.
PUBMED | CROSSREF
5. Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coman RL. Two types of murine helper T cell
clone. I. Denition according to proles of lymphokine activities and secreted proteins.
J Immunol
1986;136:2348-2357.
PUBMED | CROSSREF
6. Bettelli E, Kuchroo VK. IL-12- and IL-23-induced T helper cell subsets: birds of the same feather ock
together.
J Exp Med
2005;201:169-171.
PUBMED | CROSSREF
7. Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM, Weaver CT. Interleukin
17-producing CD4
+
eector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages.
Nat Immunol
2005;6:1123-1132.
PUBMED | CROSSREF
8. Park H, Li Z, Yang XO, Chang SH, Nurieva R, Wang YH, Wang Y, Hood L, Zhu Z, Tian Q, et al. A distinct lineage
of CD4 T cells regulates tissue inammation by producing interleukin 17.
Nat Immunol
2005;6:1133-1141.
PUBMED | CROSSREF
9. Patel DD, Kuchroo VK. Th17 cell pathway in human immunity: lessons from genetics and therapeutic
interventions.
Immunity
2015;43:1040-1051.
PUBMED | CROSSREF
10. Ouyang W, Kolls JK, Zheng Y. The biological functions of T helper 17 cell eector cytokines in
inammation.
Immunity
2008;28:454-467.
PUBMED | CROSSREF
11. Chtanova T, Tangye SG, Newton R, Frank N, Hodge MR, Rolph MS, Mackay CR. T follicular helper cells
express a distinctive transcriptional prole, reecting their role as non-Th1/Th2 eector cells that provide
help for B cells.
J Immunol
2004;173:68-78.
PUBMED | CROSSREF
12. Nurieva RI, Chung Y, Hwang D, Yang XO, Kang HS, Ma L, Wang YH, Watowich SS, Jetten AM, Tian Q, et
al. Generation of T follicular helper cells is mediated by interleukin-21 but independent of T helper 1, 2, or
17 cell lineages.
Immunity
2008;29:138-149.
PUBMED | CROSSREF
13. Kim CH, Lim HW, Kim JR, Rott L, Hillsamer P, Butcher EC. Unique gene expression program of human
germinal center T helper cells.
Blood
2004;104:1952-1960.
PUBMED | CROSSREF
T Helper Cell Differentiation and Function
https://doi.org/10.4110/in.2023.23.e4
10/15
https://immunenetwork.org
14. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated
T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance
causes various autoimmune diseases.
J Immunol
1995;155:1151-1164.
PUBMED | CROSSREF
15. Veldhoen M, Uyttenhove C, van Snick J, Helmby H, Westendorf A, Buer J, Martin B, Wilhelm C, Stockinger
B. Transforming growth factor-beta ‘reprograms’ the dierentiation of T helper 2 cells and promotes an
interleukin 9-producing subset.
Nat Immunol
2008;9:1341-1346.
PUBMED | CROSSREF
16. Dardalhon V, Awasthi A, Kwon H, Galileos G, Gao W, Sobel RA, Mitsdoerer M, Strom TB, Elyaman W,
Ho IC, et al. IL-4 inhibits TGF-β-induced Foxp3
+
T cells and, together with TGF-β, generates IL-9
+
IL-10
+
Foxp3
-
eector T cells.
Nat Immunol
2008;9:1347-1355.
PUBMED | CROSSREF
17. Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets.
Nat Rev Immunol
2011;11:723-737.
PUBMED | CROSSREF
18. Maizels RM, Pearce EJ, Artis D, Yazdanbakhsh M, Wynn TA. Regulation of pathogenesis and immunity in
helminth infections.
J Exp Med
2009;206:2059-2066.
PUBMED | CROSSREF
19. Shulman Z, Gitlin AD, Weinstein JS, Lainez B, Esplugues E, Flavell RA, Cra JE, Nussenzweig
MC. Dynamic signaling by T follicular helper cells during germinal center B cell selection.
Science
2014;345:1058-1062.
PUBMED | CROSSREF
20. Kitagawa Y, Sakaguchi S. Molecular control of regulatory T cell development and function.
Curr Opin
Immunol
2017;49:64-70.
PUBMED | CROSSREF
21. Huang CT, Workman CJ, Flies D, Pan X, Marson AL, Zhou G, Hipkiss EL, Ravi S, Kowalski J, Levitsky HI,
et al. Role of LAG-3 in regulatory T cells.
Immunity
2004;21:503-513.
PUBMED | CROSSREF
22. Paust S, Lu L, McCarty N, Cantor H. Engagement of B7 on eector T cells by regulatory T cells prevents
autoimmune disease.
Proc Natl Acad Sci U S A
2004;101:10398-10403.
PUBMED | CROSSREF
23. Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, Chen JF, Enjyoji K, Linden J, Oukka M, et
al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune
suppression.
J Exp Med
2007;204:1257-1265.
PUBMED | CROSSREF
24. Djuretic IM, Levanon D, Negreanu V, Groner Y, Rao A, Ansel KM. Transcription factors T-bet and Runx3
cooperate to activate
Ifng
and silence
Il4
in T helper type 1 cells.
Nat Immunol
2007;8:145-153.
PUBMED | CROSSREF
25. Szabo SJ, Dighe AS, Gubler U, Murphy KM. Regulation of the interleukin (IL)-12R beta 2 subunit
expression in developing T helper 1 (Th1) and Th2 cells.
J Exp Med
1997;185:817-824.
PUBMED | CROSSREF
26. Johnston RJ, Poholek AC, DiToro D, Yusuf I, Eto D, Barnett B, Dent AL, Cra J, Crotty S. Bcl6 and Blimp-1 are
reciprocal and antagonistic regulators of T follicular helper cell dierentiation.
Science
2009;325:1006-1010.
PUBMED | CROSSREF
27. Nurieva RI, Chung Y, Martinez GJ, Yang XO, Tanaka S, Matskevitch TD, Wang YH, Dong C. Bcl6 mediates
the development of T follicular helper cells.
Science
2009;325:1001-1005.
PUBMED | CROSSREF
28. Lee JB, Kim HR, Ha SJ. Immune checkpoint inhibitors in 10 years: contribution of basic research and
clinical application in cancer immunotherapy.
Immune Netw
2022;22:e2.
PUBMED | CROSSREF
29. Jung SM, Kim WU. Targeted immunotherapy for autoimmune disease.
Immune Netw
2022;22:e9.
PUBMED | CROSSREF
30. Vahedi G, Takahashi H, Nakayamada S, Sun HW, Sartorelli V, Kanno Y, O’Shea JJ. STATs shape the active
enhancer landscape of T cell populations.
Cell
2012;151:981-993.
PUBMED | CROSSREF
31. Samstein RM, Arvey A, Josefowicz SZ, Peng X, Reynolds A, Sandstrom R, Neph S, Sabo P, Kim JM, Liao
W, et al. Foxp3 exploits a pre-existent enhancer landscape for regulatory T cell lineage specication.
Cell
2012;151:153-166.
PUBMED | CROSSREF
T Helper Cell Differentiation and Function
https://doi.org/10.4110/in.2023.23.e4 11/15
https://immunenetwork.org
32. Chalmin F, Humblin E, Ghiringhelli F, Végran F. Transcriptional programs underlying Cd4 T cell
dierentiation and functions.
Int Rev Cell Mol Biol
2018;341:1-61.
PUBMED | CROSSREF
33. Christie D, Zhu J. Transcriptional regulatory networks for CD4 T cell dierentiation.
Curr Top Microbiol
Immunol
2014;381:125-172.
PUBMED | CROSSREF
34. Ciofani M, Madar A, Galan C, Sellars M, Mace K, Pauli F, Agarwal A, Huang W, Parkhurst CN, Muratet M,
et al. A validated regulatory network for Th17 cell specication.
Cell
2012;151:289-303.
PUBMED | CROSSREF
35. Yosef N, Shalek AK, Gaublomme JT, Jin H, Lee Y, Awasthi A, Wu C, Karwacz K, Xiao S, Jorgolli M, et al.
Dynamic regulatory network controlling TH17 cell dierentiation.
Nature
2013;496:461-468.
PUBMED | CROSSREF
36. Fu W, Ergun A, Lu T, Hill JA, Haxhinasto S, Fassett MS, Gazit R, Adoro S, Glimcher L, Chan S, et al.
A multiply redundant genetic switch ‘locks in’ the transcriptional signature of regulatory T cells.
Nat
Immunol
2012;13:972-980.
PUBMED | CROSSREF
37. Wilson CB, Rowell E, Sekimata M. Epigenetic control of T-helper-cell dierentiation.
Nat Rev Immunol
2009;9:91-105.
PUBMED | CROSSREF
38. Lee GR, Kim ST, Spilianakis CG, Fields PE, Flavell RA. T helper cell dierentiation: regulation by cis
elements and epigenetics.
Immunity
2006;24:369-379.
PUBMED | CROSSREF
39. Wei G, Wei L, Zhu J, Zang C, Hu-Li J, Yao Z, Cui K, Kanno Y, Roh TY, Watford WT, et al. Global
mapping of H3K4me3 and H3K27me3 reveals specicity and plasticity in lineage fate determination of
dierentiating CD4
+
T cells.
Immunity
2009;30:155-167.
PUBMED | CROSSREF
40. Levings PP, Bungert J. The human β-globin locus control region.
Eur J Biochem
2002;269:1589-1599.
PUBMED | CROSSREF
41. Lee GR, Fields PE, Grin TJ 4th, Flavell RA. Regulation of the Th2 cytokine locus by a locus control
region.
Immunity
2003;19:145-153.
PUBMED | CROSSREF
42. Koh BH, Hwang SS, Kim JY, Lee W, Kang MJ, Lee CG, Park JW, Flavell RA, Lee GR. Th2 LCR is essential
for regulation of Th2 cytokine genes and for pathogenesis of allergic asthma.
Proc Natl Acad Sci U S A
2010;107:10614-10619.
PUBMED | CROSSREF
43. Hwang SS, Kim YU, Lee S, Jang SW, Kim MK, Koh BH, Lee W, Kim J, Souabni A, Busslinger M, et
al. Transcription factor YY1 is essential for regulation of the Th2 cytokine locus and for Th2 cell
dierentiation.
Proc Natl Acad Sci U S A
2013;110:276-281.
PUBMED | CROSSREF
44. Hwang SS, Jang SW, Lee KO, Kim HS, Lee GR. RHS6 coordinately regulates the Th2 cytokine genes by
recruiting GATA3, SATB1, and IRF4.
Allergy
2017;72:772-782.
PUBMED | CROSSREF
45. Lee GR, Spilianakis CG, Flavell RA. Hypersensitive site 7 of the TH2 locus control region is essential
for expressing TH2 cytokine genes and for long-range intrachromosomal interactions.
Nat Immunol
2005;6:42-48.
PUBMED | CROSSREF
46. Williams A, Lee GR, Spilianakis CG, Hwang SS, Eisenbarth SC, Flavell RA. Hypersensitive site 6 of the Th2
locus control region is essential for Th2 cytokine expression.
Proc Natl Acad Sci U S A
2013;110:6955-6960.
PUBMED | CROSSREF
47. Spilianakis CG, Flavell RA. Long-range intrachromosomal interactions in the T helper type 2 cytokine
locus.
Nat Immunol
2004;5:1017-1027.
PUBMED | CROSSREF
48. Spilianakis CG, Lalioti MD, Town T, Lee GR, Flavell RA. Interchromosomal associations between
alternatively expressed loci.
Nature
2005;435:637-645.
PUBMED | CROSSREF
49. Kim LK, Esplugues E, Zorca CE, Parisi F, Kluger Y, Kim TH, Galjart NJ, Flavell RA. Oct-1 regulates IL-17
expression by directing interchromosomal associations in conjunction with CTCF in T cells.
Mol Cell
2014;54:56-66.
PUBMED | CROSSREF
T Helper Cell Differentiation and Function
https://doi.org/10.4110/in.2023.23.e4
12/15
https://immunenetwork.org
50. Hadjur S, Williams LM, Ryan NK, Cobb BS, Sexton T, Fraser P, Fisher AG, Merkenschlager M. Cohesins form
chromosomal cis-interactions at the developmentally regulated IFNG locus.
Nature
2009;460:410-413.
PUBMED | CROSSREF
51. Sekimata M, Pérez-Melgosa M, Miller SA, Weinmann AS, Sabo PJ, Sandstrom R, Dorschner MO,
Stamatoyannopoulos JA, Wilson CB. CCCTC-binding factor and the transcription factor T-bet orchestrate
T helper 1 cell-specic structure and function at the interferon-gamma locus.
Immunity
2009;31:551-564.
PUBMED | CROSSREF
52. Murphy E, Shibuya K, Hosken N, Openshaw P, Maino V, Davis K, Murphy K, O’Garra A. Reversibility of T
helper 1 and 2 populations is lost aer long-term stimulation.
J Exp Med
1996;183:901-913.
PUBMED | CROSSREF
53. Grogan JL, Mohrs M, Harmon B, Lacy DA, Sedat JW, Locksley RM. Early transcription and silencing of
cytokine genes underlie polarization of T helper cell subsets.
Immunity
2001;14:205-215.
PUBMED | CROSSREF
54. Ballesteros-Tato A, Randall TD, Lund FE, Spolski R, Leonard WJ, León B. T follicular helper cell plasticity
shapes pathogenic T helper 2 cell-mediated immunity to inhaled house dust mite.
Immunity
2016;44:259-273.
PUBMED | CROSSREF
55. Zhou L, Chong MM, Littman DR. Plasticity of CD4
+
T cell lineage dierentiation.
Immunity
2009;30:646-655.
PUBMED | CROSSREF
56. Hirota K, Duarte JH, Veldhoen M, Hornsby E, Li Y, Cua DJ, Ahlfors H, Wilhelm C, Tolaini M, Menzel U, et
al. Fate mapping of IL-17-producing T cells in inammatory responses.
Nat Immunol
2011;12:255-263.
PUBMED | CROSSREF
57. Bending D, De la Peña H, Veldhoen M, Phillips JM, Uyttenhove C, Stockinger B, Cooke A. Highly puried
Th17 cells from BDC2.5NOD mice convert into Th1-like cells in NOD/SCID recipient mice.
J Clin Invest
2009;119:565-572.
PUBMED | CROSSREF
58. Muranski P, Borman ZA, Kerkar SP, Klebano CA, Ji Y, Sanchez-Perez L, Sukumar M, Reger RN, Yu
Z, Kern SJ, et al. Th17 cells are long lived and retain a stem cell-like molecular signature.
Immunity
2011;35:972-985.
PUBMED | CROSSREF
59. Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, Cua DJ, Littman DR. The orphan
nuclear receptor RORγt directs the dierentiation program of proinammatory IL-17
+
T helper cells.
Cell
2006;126:1121-1133.
PUBMED | CROSSREF
60. Okada S, Markle JG, Deenick EK, Mele F, Averbuch D, Lagos M, Alzahrani M, Al-Muhsen S, Halwani
R, Ma CS, et al. Impairment of immunity to
Candida
and
Mycobacterium
in humans with bi-allelic RORC
mutations.
Science
2015;349:606-613.
PUBMED | CROSSREF
61. Koch MA, Tucker-Heard G, Perdue NR, Killebrew JR, Urdahl KB, Campbell DJ. The transcription factor
T-bet controls regulatory T cell homeostasis and function during type 1 inammation.
Nat Immunol
2009;10:595-602.
PUBMED | CROSSREF
62. Wan YY, Flavell RA. Regulatory T-cell functions are subverted and converted owing to attenuated Foxp3
expression.
Nature
2007;445:766-770.
PUBMED | CROSSREF
63. Zheng Y, Chaudhry A, Kas A, deRoos P, Kim JM, Chu TT, Corcoran L, Treuting P, Klein U, Rudensky AY.
Regulatory T-cell suppressor program co-opts transcription factor IRF4 to control T
H
2 responses.
Nature
2009;458:351-356.
PUBMED | CROSSREF
64. Chaudhry A, Rudra D, Treuting P, Samstein RM, Liang Y, Kas A, Rudensky AY. CD4
+
regulatory T cells
control T
H
17 responses in a Stat3-dependent manner.
Science
2009;326:986-991.
PUBMED | CROSSREF
65. Roy S, Rizvi ZA, Awasthi A. Metabolic checkpoints in dierentiation of helper T cells in tissue
inammation.
Front Immunol
2019;9:3036.
PUBMED | CROSSREF
66. Almeida L, Dhillon-LaBrooy A, Carriche G, Berod L, Sparwasser T. CD4
+
T-cell dierentiation and
function: unifying glycolysis, fatty acid oxidation, polyamines NAD mitochondria.
J Allergy Clin Immunol
2021;148:16-32.
PUBMED | CROSSREF
6 7. Yang W, Yu T, Cong Y. CD4
+
T cell metabolism, gut microbiota, and autoimmune diseases: implication in
precision medicine of autoimmune diseases.
Precis Clin Med
2022;5:pbac018.
PUBMED | CROSSREF
T Helper Cell Differentiation and Function
https://doi.org/10.4110/in.2023.23.e4 13/15
https://immunenetwork.org
68. Xu K, Yin N, Peng M, Stamatiades EG, Chhangawala S, Shyu A, Li P, Zhang X, Do MH, Capistrano KJ, et
al. Glycolytic ATP fuels phosphoinositide 3-kinase signaling to support eector T helper 17 cell responses.
Immunity
2021;54:976-987.e7.
PUBMED | CROSSREF
69. Shi LZ, Wang R, Huang G, Vogel P, Neale G, Green DR, Chi H. HIF1alpha-dependent glycolytic
pathway orchestrates a metabolic checkpoint for the dierentiation of TH17 and Treg cells.
J Exp Med
2011;208:1367-1376.
PUBMED | CROSSREF
70. Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, Mason EF, Sullivan SA, Nichols AG,
Rathmell JC. Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for
eector and regulatory CD4
+
T cell subsets.
J Immunol
2011;186:3299-3303.
PUBMED | CROSSREF
71. Wang R, Dillon CP, Shi LZ, Milasta S, Carter R, Finkelstein D, McCormick LL, Fitzgerald P, Chi H, Munger
J, et al. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation.
Immunity
2011;35:871-882.
PUBMED | CROSSREF
72. Preston GC, Sinclair LV, Kaskar A, Hukelmann JL, Navarro MN, Ferrero I, MacDonald HR, Cowling VH,
Cantrell DA. Single cell tuning of Myc expression by antigen receptor signal strength and interleukin-2 in
T lymphocytes.
EMBO J
2015;34:2008-2024.
PUBMED | CROSSREF
73. Man K, Kallies A. Synchronizing transcriptional control of T cell metabolism and function.
Nat Rev
Immunol
2015;15:574-584.
PUBMED | CROSSREF
74. McNamee EN, Korns Johnson D, Homann D, Clambey ET. Hypoxia and hypoxia-inducible factors as
regulators of T cell development, dierentiation, and function.
Immunol Res
2013;55:58-70.
PUBMED | CROSSREF
75. Dang EV, Barbi J, Yang HY, Jinasena D, Yu H, Zheng Y, Bordman Z, Fu J, Kim Y, Yen HR, et al. Control of
T
H
17/T
reg
balance by hypoxia-inducible factor 1.
Cell
2011;146:772-784.
PUBMED | CROSSREF
76. Doedens AL, Phan AT, Stradner MH, Fujimoto JK, Nguyen JV, Yang E, Johnson RS, Goldrath AW. Hypoxia-
inducible factors enhance the eector responses of CD8
+
T cells to persistent antigen.
Nat Immunol
2013;14:1173-1182.
PUBMED | CROSSREF
77. Phan AT, Doedens AL, Palazon A, Tyrakis PA, Cheung KP, Johnson RS, Goldrath AW. Constitutive
glycolytic metabolism supports CD8
+
T cell eector memory dierentiation during viral infection.
Immunity
2016;45:1024-1037.
PUBMED | CROSSREF
78. Clever D, Roychoudhuri R, Constantinides MG, Askenase MH, Sukumar M, Klebano CA, Eil RL,
Hickman HD, Yu Z, Pan JH, et al. Oxygen sensing by T cells establishes an immunologically tolerant
metastatic niche.
Cell
2016;166:1117-1131.e14.
PUBMED | CROSSREF
79. Delgoe GM, Kole TP, Zheng Y, Zarek PE, Matthews KL, Xiao B, Worley PF, Kozma SC, Powell JD. The
mTOR kinase dierentially regulates eector and regulatory T cell lineage commitment.
Immunity
2009;30:832-844.
PUBMED | CROSSREF
80. Delgoe GM, Pollizzi KN, Waickman AT, Heikamp E, Meyers DJ, Horton MR, Xiao B, Worley PF, Powell
JD. The kinase mTOR regulates the dierentiation of helper T cells through the selective activation of
signaling by mTORC1 and mTORC2.
Nat Immunol
2011;12:295-303.
PUBMED | CROSSREF
81. Lee K, Gudapati P, Dragovic S, Spencer C, Joyce S, Killeen N, Magnuson MA, Boothby M. Mammalian
target of rapamycin protein complex 2 regulates dierentiation of Th1 and Th2 cell subsets via distinct
signaling pathways.
Immunity
2010;32:743-753.
PUBMED | CROSSREF
82. Shrestha S, Yang K, Guy C, Vogel P, Neale G, Chi H. T
reg
cells require the phosphatase PTEN to restrain T
H
1
and T
FH
cell responses.
Nat Immunol
2015;16:178-187.
PUBMED | CROSSREF
83. Huynh A, DuPage M, Priyadharshini B, Sage PT, Quiros J, Borges CM, Townamchai N, Gerriets VA,
Rathmell JC, Sharpe AH, et al. Control of PI(3) kinase in T
reg
cells maintains homeostasis and lineage
stability.
Nat Immunol
2015;16:188-196.
PUBMED | CROSSREF
T Helper Cell Differentiation and Function
https://doi.org/10.4110/in.2023.23.e4
14/15
https://immunenetwork.org
84. Hardie DG. AMP-activated protein kinase: an energy sensor that regulates all aspects of cell function.
Genes Dev
2011;25:1895-1908.
PUBMED | CROSSREF
85. Meares GP, Qin H, Liu Y, Holdbrooks AT, Benveniste EN. AMP-activated protein kinase restricts IFN-γ
signaling.
J Immunol
2013;190:372-380.
PUBMED | CROSSREF
86. Gaublomme JT, Yosef N, Lee Y, Gertner RS, Yang LV, Wu C, Pandol PP, Mak T, Satija R, Shalek AK, et al.
Single-cell genomics unveils critical regulators of Th17 cell pathogenicity.
Cell
2015;163:1400-1412.
PUBMED | CROSSREF
87. Karmaus PW, Chen X, Lim SA, Herrada AA, Nguyen TM, Xu B, Dhungana Y, Rankin S, Chen W,
Rosencrance C, et al. Metabolic heterogeneity underlies reciprocal fates of T
H
17 cell stemness and
plasticity.
Nature
2019;565:101-105.
PUBMED | CROSSREF
88. Tortola L, Jacobs A, Pohlmeier L, Obermair FJ, Ampenberger F, Bodenmiller B, Kopf M. High-
dimensional T helper cell proling reveals a broad diversity of stably committed eector states and
uncovers interlineage relationships.
Immunity
2020;53:597-613.e6.
PUBMED | CROSSREF
89. Kiner E, Willie E, Vijaykumar B, Chowdhary K, Schmutz H, Chandler J, Schnell A, Thakore PI, LeGros G,
Mostafavi S, et al. Gut CD4
+
T cell phenotypes are a continuum molded by microbes, not by T
H
archetypes.
Nat Immunol
2021;22:216-228.
PUBMED | CROSSREF
90. nnberg T, Svensson V, James KR, Fernandez-Ruiz D, Sebina I, Montandon R, Soon MS, Fogg LG, Nair
AS, Liligeto U, et al. Single-cell RNA-seq and computational analysis using temporal mixture modelling
resolves Th1/T fate bifurcation in malaria.
Sci Immunol
2017;2:eaal2192.
PUBMED | CROSSREF
T Helper Cell Differentiation and Function
https://doi.org/10.4110/in.2023.23.e4 15/15
https://immunenetwork.org