Abstract
Hypoxia‐inducible factor‐1 (HIF‐1) is a heterodimeric (HIF‐1α/ HIF‐1β) transcription factor in which the oxygen‐sensitive HIF‐1α subunit regulates gene transcription to mediate adaptive tissue responses to hypoxia. HIF‐1 is a key mediator in both regulatory and pathogenic immune responses, because ongoing inflammation in localized tissues causes increased oxygen consumption and consequent hypoxia within the inflammatory lesions. In autoimmune diseases, HIF‐1 plays complex and divergent roles within localized inflammatory lesions by orchestrating a critical immune interplay sponsoring the pathogenesis of the disease. In this review, we have summarized the role of HIF‐1 in lymphoid and myeloid immunomodulation in autoimmune diseases. HIF‐1 drives inflammation by controlling the Th17/Treg/Tr1 balance through the tipping of the differentiation of CD4+ T cells in favour of pro‐inflammatory Th17 cells while suppressing the development of anti‐inflammatory Treg/Tr1 cells. On the other hand, HIF‐1 plays a protective role by facilitating the expression of anti‐inflammatory cytokine IL‐10 in and expansion of CD1dhiCD5+ B cells, known as regulatory B cells or B10 cells. Apart from lymphoid cells, HIF‐1 also controls the activation of macrophages, neutrophils and dendritic cells, thus eventually further influences the activation and development of effector/regulatory T cells by facilitating the creation of a pro/anti‐inflammatory microenvironment within the autoinflammatory lesions. Based on the critical immunomodulatory roles that HIF‐1 plays, this master transcription factor seems to be a potent druggable target for the treatment of autoimmune diseases.
Keywords: autoimmune diseases, HIF‐1, hypoxia, lymphocytes, myeloid cells
Hypoxia‐inducible factor‐1 is a master transcription factor, which regulates different gene transcriptions in response to hypoxia following an oxygen‐sensitive mechanism. HIF‐1 modulates lymphoid and myeloid immune responses in autoimmune diseases in a divergent manner. HIF‐1 drives autoimmune inflammation by controlling divergent pro/anti‐inflammatory function of different immune cells located in the inflammatory lesions.

Abbreviations
- AHR
aryl hydrocarbon receptor
- B10
IL‐10‐producing regulatory B cells
- C‐MAF
C‐musculoaponeurotic fibrosarcoma oncogene
- E3L
ubiquitin ligase 3
- EAE
experimental autoimmune encephalomyelitis
- eATP
extracellular adenosine triphosphate
- FOXP3
forkhead box P3
- HIF‐1
hypoxia‐inducible factor‐1
- IGM
insulin growth factor
- iNOS
inducible nitric oxide synthase
- MIP‐β
macrophage inflammatory protein‐β
- NF‐κB
nuclear factor kappa‐light‐chain‐enhancer of activated B cells
- NO
nitric oxide
- P300
P300 transcriptional coactivator protein
- PHDs
prolyl hydroxylases
- pVHL
von Hippel–Lindau tumour suppressor protein
- RNS
reactive nitrogen species
- RORγt
retinoic‐acid‐receptor‐related orphan nuclear receptor gamma
- ROS
reactive oxygen species
- SOCS3
suppressor of cytokine signalling 3
- STAT3
signal transducer and activator of transcription 3
- STAT4
signal transducer and activator of transcription 4
- Teff
effector T cells
- Th1
T‐helper 1 cells
- Th17
T‐helper 17 cells
- Tr1
type 1 regulatory T cells
- Treg
regulatory T cells
INTRODUCTION
Hypoxia‐inducible factor‐1 (HIF‐1) is a heterodimeric transcription factor consisting of HIF‐1α and HIF‐1β. HIF‐1β is constitutively expressed, whereas HIF‐1α is an oxygen sensor and its availability is inversely regulated by cellular oxygen tension [1]. Under normoxic condition, HIF‐1α is hydroxylated at specific proline residues by oxygen‐dependent prolyl hydroxylases (PHDs). Prolyl hydroxylation signals for ubiquitin proteasome‐mediated degradation of HIF‐α. von Hippel–Lindau tumour suppressor protein (pVHL), an E3 ubiquitin ligase, recognizes and binds to hydroxylated HIF‐1‐α [2]. pVHL, bound with HIF‐1α, acts as the substrate recognition component of E3 ubiquitin ligase complex and possesses ubiquitin ligase activity. This complex leads to the ubiquitination and subsequent proteasomal degradation of HIF‐1α. But under hypoxic condition, activity of PHDs is suppressed due to the reduced oxygen availability, resulting in stabilization and accumulation of HIF‐1α, translocation of HIF‐1α into the nucleus and heterodimerization with HIF‐1β. Heterodimerized HIF‐1 then binds to hypoxia‐responsive elements (HREs) as a transcription factor and activates the transcription of target genes [3] (Figure 1).
FIGURE 1.

Hypoxia induces stabilization, accumulation and nuclear translocation of HIF‐1α and therefore induces the transcription of HIF‐1 target genes. The availability of HIF‐1α in the cytoplasm is controlled by cellular oxygen tension. Under normoxic condition, HIF‐1α undergoes hydroxylation at specific proline residues by PHDs. Prolyl‐hydroxylated HIF‐1α is recognized by pVHL. Binding of hydroxylated HIF‐1α by pVHL leads to the ubiquitination of HIF‐1α. Polyubiquitinated HIF‐1α undergoes proteasomal degradation. Under hypoxic condition, prolyl hydroxylation of HIF‐1α is suppressed, leading to its stabilization, accumulation and nuclear translocation. Within nucleus, HIF‐1α dimerizes with HIF‐1β, binds to HRE, interacts with P300 and induces the transcription of HIF‐1 target genes. HIF‐1, hypoxia‐inducible factor‐1; HRE: hypoxia‐responsive element; P300, P300 transcriptional coactivator protein; PHDs, prolyl hydroxylases; pVHL, von Hippel–Lindau tumour suppressor protein
Hypoxia is defined as a condition with reduced oxygen availability relative to oxygen demand and is contingent upon the pathophysiological circumstance of the afflicted organ or tissue [4]. Hypoxia is intricately related to inflammation, and these two processes mutually contribute to each other and act by reinforcing mechanisms [5, 6, 7]. Hypoxia‐elicited inflammation has been implicated in a wide range of disease conditions [7]. And vice versa, inflammation‐induced hypoxia or ‘inflammatory hypoxia’ reflects the well‐established concept that ongoing inflammation causes increased oxygen consumption, which results in localized tissue hypoxia within inflamed tissues [5, 6]. Multiple mechanisms contribute to the development of hypoxia in inflammatory lesions. One important mechanism is the excessive consumption of available oxygen by the immigrating myeloid cells and activated lymphocytes within the inflammatory lesions [5, 8]. Myeloid cells consume substantial quantities of nutrients, energy and oxygen to accomplish the processes of migration to the sites of inflammation, phagocytosis and release of reactive oxygen species (ROS) [9, 10, 11]. In the case of lymphocytes, clonal expansion in inflammatory sites is an energy‐intensive process, which is predominantly dependent on oxidative phosphorylation during early phases of inflammation [5, 12]. Another important mechanism of elevated oxygen consumption in inflammatory sites is the increased generation of ROS and reactive nitrogen species (RNS). Elevated level of nitric oxide (NO) is generated by enhanced expression of different isoforms of NO synthase in neuron, endothelial cells and immune cells [13]. Increased generation of superoxide (O2 −) by activated neutrophils and macrophages in inflammatory sites can also result in diminished availability of molecular oxygen [14, 15, 16]. NO and O2 − generated in inflammatory lesions react to produce the highly toxic oxidant peroxynitrite (ONOO−) [17]. Excessive generation of NO and ONOO− contributes to the random damage of surrounding healthy tissue, leading to the exacerbation of inflammation [13, 17, 18]. Hypoxia developed within the inflammatory lesions, in turn, facilitates the stabilization of HIF‐1α and thus leads to the activation of transcription of target genes [3].
Autoimmune diseases are characterized by tissue destruction caused by misguided autoreactive cell‐mediated or humoral immune responses [19]. Because hypoxia within the inflammatory lesions is a sustained microenvironmental feature in autoimmune diseases, HIF‐1 has been implicated as a pathogenic mediator that enables persistence of inflammation despite the unfavourable hypoxic condition [20, 21, 22]. Indeed, a growing body of evidences revealed that HIF‐1α mediates a critical role in inflammation and pathogenesis of autoimmune diseases, including rheumatoid arthritis, inflammatory bowel disease, psoriasis, type 1 diabetes mellitus, systemic sclerosis and multiple sclerosis [23, 24, 25, 26, 27, 28]. Following inflammation‐elicited hypoxia within the inflammatory lesions, the level of stabilized HIF‐1 is increased, which facilitates the adaptation of the inflamed tissue to the hypoxic microenvironment through the transcriptional activation of target genes [29, 30, 31, 32]. Target genes of HIF‐1 vary widely depending on the cell type, tissue and pathophysiological context. At present, more than 100 direct target genes of HIF‐1 have been identified, which subserve a diverse function in inflammation, angiogenesis, tumorigenesis, energy metabolism, cell survival, differentiation, proliferation and apoptosis [6, 33, 34]. In conjunction with oxygen homeostasis and hypoxia adaptation, HIF‐1 performs an immunomodulatory role, which is critically important for directing the course of progression of autoimmune diseases. Immunomodulatory function of HIF‐1 in autoimmune diseases is very divergent and dependent on immune cell lineages and is intricately connected to other mediators of hypoxic inflammation, for instance, cytokines, hormones such as insulin or insulin‐like growth factors (IGF‐1 and IGF‐2), vasoactive peptides, such as angiotensin II, and availability of NO [5, 35]. This review illustrates the multifaceted role of HIF‐1 in modulating the lineage‐specific immune responses of autoimmune diseases.
LYMPHOCYTES
A safe route of inflammatory response of T cells requires the triggering of protective inflammation to eliminate infection, resolution of inflammatory phase after the eradication of invading elements, and containment of dysregulated or overaggressive immune sequelae. However, many mechanisms may contribute to autoimmune pathogenesis, including molecular mimicry, chronic/latent recurrent infection or post‐infectious immune dysregulation. An uncontained self‐aggressive inflammatory response of T cell can result in autoimmune tissue damage. Among different subsets of T cells, Th17 is the most prominent effector T cell in orchestrating T‐cell‐mediated autoimmune diseases, for example multiple sclerosis and experimental autoimmune encephalomyelitis (EAE) (a widely used animal model of multiple sclerosis), collagen‐induced arthritis, inflammatory bowel disease, uveitis and type 1 diabetes [36, 37, 38, 39] On the other hand, proliferation and autoimmune responses of effector T cells, including Th17 cells, are suppressed by anti‐inflammatory regulatory T (Treg) cells [40, 41, 42]. Therefore, a fine balance is maintained between Th17 and Treg cells during homeostasis, and interruption of this delicate balance can potentially threaten with the development of autoimmune diseases.
In the context of autoimmunity, non‐pathogenic Th17 cells are induced from naïve T cells by IL‐6 and TGF‐β, whereas the pathogenic Th17 differentiation programme requires IL‐23. Conversely, Treg cells are induced from naïve T cells by TGF‐β and IL‐2 and undergo clonal expansion upon continued exposure to IL‐2 [40, 41, 43, 44, 45, 46]. The differentiation programmes of both Th17 and Treg subsets have a common requirement for TGF‐β, which drives enhanced expression of the signature transcription factors RORγt and FOXP3, respectively. In addition to TGF‐β, differentiation of Th17 cells requires an additional signal from IL‐6 pathway. Therefore, differentiation of either Th17 or Treg cells from naïve T cells becomes dominant depending on the relative abundance of IL‐6 and TGF‐β within the inflammatory lesion [46, 47]. Given that, TGF‐β induces the coexpression of RORγt and Foxp3 in naïve T cells [48, 49]. But, in the presence of high level of TGF‐β and absence of IL‐6, Foxp3 directly interacts, at least in part, with the RORγt and antagonizes its ability to bind the DNA, which eventually directs the differentiation of T cells away from Th17 transcriptional programme and towards the Treg subtype. Nevertheless, in the presence of IL‐6 and low level of TGF‐β, signal transducer and activator of transcription 3 (STAT3) becomes activated, which allows the transcriptional activity of RORγt by overcoming the inhibition by Foxp3. Therefore, differentiation of Th17 is dependent on a sustained and unopposed transcriptional activity of STAT3 and RORγt [47, 50]. Enhanced activation of STAT3, however, may not be sufficient enough, by itself, to tilt the delicate balance between Treg and Th17 towards pathogenic Th17. Therefore, a simultaneous downregulation of Foxp3 coupled with sustained expression of RORγt is also critical for pathogenic Th17 differentiation.
In the hypoxic milieu of inflammatory lesion, level of available HIF‐1 within the T cells is increased, which could be contributed both by the enhanced transcription of HIF‐1 through activated STAT3 under inflammatory condition and by the stabilization of HIF‐1 under low oxygen tension [46, 51]. Accumulated HIF‐1 plays a three‐pronged role in the skewed differentiation of Th17 cells in the inflammatory lesions (Figure 2). First, HIF‐1 directly binds to the hypoxia‐responsive element (HRE) at the promoter of RORγt and induces its expression in co‐operation with STAT3. Second, HIF‐1 associates with RORγt at the promoter of Th17 genes and recruits transcriptional coactivator P300 to the transcription complex, resulting in the generation of a permissive chromatin structure. Third, HIF‐1 suppresses the differentiation of Treg by directly interacting Foxp3 and facilitating its degradation via PHD/VHL/ubiquitin‐mediated proteasomal degradation, the same ubiquitin ligase system responsible for the degradation of HIF‐1α itself. Therefore, HIF‐1 transcriptionally activates the genetic programme of Th17 subset while repressing the Treg programme in the same time. In the mouse model of EAE, CD4+ T‐cell‐specific depletion of HIF‐1 results in a deficiency of IL‐17 production and an increased number of Treg cells in the central nervous system, which culminates in the attenuation disease [46]. Taken together, HIF‐1 facilitates the transcriptional programme of Th17, pushing the population ratio of Th17/Treg towards Th17 subset. In addition to the induction of RORγt transcriptional programme, HIF‐1 facilitates the metabolic switch from oxidative phosphorylation to glycolytic pathway as the primary generator of energy by inducing an increased expression of glycolytic enzymes, which reinforces the differentiation of Th17 cells over Treg cells [52, 53].
FIGURE 2.

HIF‐1 modulates the differentiation of T cells towards pathogenic Th17/Th1 or regulatory Treg/Tr1 cells depending on cytokine milieu within the hypoxic autoinflammatory lesions. The relative abundance of IL‐6, IL‐12, TGF‐β and IL‐27 dictates the differentiation of Th17, Th1, Treg or Tr1 cells. TGF‐β induces the expression of both FOXP3 and RORγt. In the absence of IL‐6, FOXP3 antagonizes the binding of RORγt to the DNA, leading to the activation of T‐cell transcriptional programme towards Treg cells. But, in the presence of IL‐6 and low level of TGF‐β, IL‐6 pathway‐activated STAT3 induces the expression of RORγt, which forms transcriptional complex with P300, HIF‐1 and STAT3 leading to the expression of IL‐17. In the presence of IL‐27, T cells become IL‐10‐producing Tr1. IL‐27 induces the expression of AHR through the activation of STAT3 pathway. AHR together with C‐MAF induces the expression of IL‐10 and IL‐21. IL‐21 promotes the stability and growth of Tr1 through autocrine signalling. AHR together with STAT3 also induces the expression of ectonucleotidase CD39. CDC9 degrades ATP, a suppressor of Tr1, into AMP on the plasma membrane. In the presence of IL‐12, T cells become IFN‐γ‐producing Th1 cells through the activation of STAT4 pathway. Hypoxia within the inflammatory lesions leads to the accumulation of HIF‐1, also induced by IL‐6‐activated STAT3, which interacts with FOXP3 or AHR and facilitates their degradation via pVHL‐E3L‐mediated proteasomal degradation, suppressing the differentiation of Treg or Tr1 cells. HIF1 inhibits the expression of STAT4 and therefore suppresses the development of IFN‐γ‐producing Th1 cells. HIF‐1 also inhibits the expression of SOCS3, which forms a feedback inhibitory loop with STAT3 and therefore promotes the differentiation of Th17 cells by restricting the suppression of STAT3 activation. HIF‐1 also induces the expression of glycolytic enzymes, which promotes the differentiation of Th17 cells over Treg cells by facilitating the metabolic switch from oxidative phosphorylation to glycolytic pathway. HIF‐1 not only enhances but also inhibits the differentiation of Th17 by inducing the expression of iNOS, which suppresses the transcriptional activity of RORγt by NO‐mediated nitration. AHR, arylhydrocarbon receptor; AMP, adenosine monophosphate; ATP, adenosine triphosphate; CD39, cluster of differentiation 39; C‐MAF, c‐musculoaponeurotic fibrosarcoma oncogene; E3L, ubiquitin ligase 3; FOXP3, forkhead box P3; HIF‐1, hypoxia‐inducible factor‐1; IFN‐γ, interferon gamma; IL‐10, interleukin‐10; IL‐12, interleukin‐12; IL‐21, interleukin‐21; IL‐27, interleukin‐27; IL‐6, interleukin‐6; iNOS, inducible nitric oxide synthase; NO, nitric oxide; P300, P300 transcriptional coactivator protein; pVHL, von Hippel–Lindau tumour suppressor protein; RORγt, retinoic‐acid‐receptor‐related orphan nuclear receptor gamma; SOCS3, suppressor of cytokine signalling 3; STAT3, signal transducer and activator of transcription 3; STAT4, signal transducer and activator of transcription 4; TGF‐β, transforming growth factor‐beta; Th1, T‐helper 1 cells; Th17, T‐helper 17 cells; Tr1, type 1 regulatory T cells; Treg, regulatory T cells
Contrary to the Th17‐skewing function, HIF‐1 may also antagonize the differentiation of Th17 cells. Yang and colleagues reported that T‐cell‐derived inducible nitric oxide synthase (iNOS), an well‐recognized target gene of HIF‐1, suppresses the IL‐17 promoter activation of T cells in vitro through the nitration of tyrosine residues (particularly Tyr346 and Tyr359) in RORᵞt, leading to the impaired differentiation and proliferation of Th17 cells [54, 55]. Moreover, T‐cell‐specific deficiency of iNOS significantly increases the percentage of IL‐17‐producing cells in the colon and exacerbates the disease in murine colitis [54]. Therefore, endogenous NO‐mediated suppression of Th17 cells contradicts the hypoxia‐induced pro‐inflammatory function of HIF‐1 in T cells in mediating the autoimmune diseases. Not only in Th17 cells but also in Treg cells, the contradictory role of HIF‐1 has also been reported. Clambey and colleagues showed that HIF‐1α, under hypoxia as an environmental cue of inflamed microenvironment, selectively induces the transcription of Foxp3 by directly binding to a single consensus hypoxia‐responsive element (HRE) within the promoter of Foxp3 gene and therefore promotes the proliferation and relative abundance of Treg cells in vitro [56]. Clambey et al. also showed that HIF‐1α is required for optimal function of Treg cells and HIF‐1α‐deficient Treg cells are defective in controlling T‐cell‐mediated colitis. Additional research is needed to understand how these opposing functions of HIF‐1 in T cells are fine‐tuned in the pro‐inflammatory setting of an autoimmune disease.
In comparison with Th17 cells, IFN‐γ‐producing pathogenic Th1 cells are less prominent in driving the pathology of tissue‐specific autoimmune diseases [57]. Shehade et al showed that HIF‐1 has an important role in restricting the function of Th1 cells [58]. Under hypoxia, HIF‐1 inhibits the transcription of STAT4, a key transcription factor mostly activated by pro‐inflammatory cytokine IL‐12 and required for the development and function of Th1 cells. Concurrently, HIF‐1 also inhibits the transcription of suppressor of cytokine signalling 3 (SOCS3), which is a major negative feedback regulator of STAT3 required for the development and function of Th17 cells [58, 59, 60, 61]. Therefore, it is clear that HIF‐1 favours the differentiation and function of pathogenic Th17 cells over Th1 cells. However, this issue requires further investigation to have a better picture of how HIF‐1 restricts the differentiation and function of Th1 cells in a complex blend of cytokines within autoimmune lesions (Figure 2).
In addition to Treg cells, type 1 regulatory T (Tr1) cells are another important CD4+ anti‐inflammatory player in the modulation of autoimmune diseases. Tr1 cells are Foxp3− and play a non‐redundant role in controlling autoimmune inflammation by producing IL‐10 [62]. Decreased numbers and impaired function of Tr1 cells have been documented in a variety of autoimmune diseases [63]. IL‐27 is the key cytokine for Tr1 polarization of naïve CD4+ T cells, which is produced by different antigen‐presenting cells including dendritic cells, monocytes and macrophages [64]. Upon interaction with IL‐27R, IL‐27 activates STAT3 through Janus kinase (JAK)/STAT pathway. When activated, STAT3 upregulates the expression of aryl hydrocarbon receptor (AHR) [65, 66]. AHR synergizes with c‐musculoaponeurotic fibrosarcoma oncogene (C‐MAF), another transcription factor induced by IL‐27, to promote the transcription of IL‐10 and IL‐21 [67]. Under the influence of IL‐27, IL‐21 acts as an autocrine growth factor for Tr1 cells [68]. AHR, together with STAT3, also induces the expression of plasma membrane ectonucleotidase CD39. CD39 translocates to the membrane and degrades extracellular ATP (eATP), a danger signal released by inflamed and dying tissue. eATP induces the differentiation of Th17 cells by engaging purinergic P2 receptors, but inhibits the differentiation of Tr1 through P2X7 receptor signalling [66, 69]. Therefore, by hydrolysing eATP, CD39 restricts eATP‐mediated suppression of Tr1 differentiation. HIF‐1α has differing roles in Tr1 differentiation. Stabilized HIF‐1α promotes AHR ubiquitination and its degradation by proteasome. Therefore, under hypoxic condition, HIF‐1α interferes with AHR‐dependent signalling and therefore restricts the differentiation of Tr1 cells and indirectly promotes the differentiation of Th17 cells [66, 70]. On the other hand, HIF‐1α facilitates the metabolic reprogramming of Tr1 cells at the early stage of differentiation by inducing the expression of glycolytic genes, which is similar to that of Th17 cells and different from the oxidative phosphorylation associated with Foxp3+ Treg cells [66]. This HIF‐1α‐induced early‐stage metabolic reprogramming could draw a diverging line between the developmental pathways of naïve T cells towards Tr1 or Treg cells under the influence of appropriate anti‐inflammatory cytokine signalling (Figure 2).
Most of the current literature regarding the role of HIF‐1 in autoimmune diseases is overwhelmingly focused on CD4+ T cells. However, in a recent study, Doedens and colleagues reported that HIF‐1 induces the cytolytic functions of CD8+ T cells (e.g. release of cytotoxic granules containing granzymes B and perforin) in the context of persistent viral and tumour antigens, which results in induced apoptosis of host cells [71]. Therefore, the plausibility of a HIF‐1‐induced and CD8+ T‐cell‐mediated tissue destruction within the hypoxic microenvironment of pro‐inflammatory lesions in autoimmune diseases cannot be ruled out.
Although numerous studies have delineated the role of HIF‐1 in T cells, the function of HIF‐1 in B cells in relation to autoimmune diseases is yet to be completely understood. However, emerging interest has been focused on the role of HIF‐1 in B cells, with a particular interest in IL‐10‐producing CD1dhi CD5+ B cells (known as regulatory B cells or B10 cells), because of their anti‐inflammatory functions in autoimmune diseases. Recently, Meng and colleagues illustrated the function of HIF‐1 in B cells implicated in the pathogenesis of collagen‐induced arthritis and EAE. In activated B cells, particularly CD1dhiCD5+ B cells, HIF‐1 level increases, which synergizes with STAT3 to induce the expression of anti‐inflammatory cytokine IL‐10 that suppresses pathogenic T cells. In the absence of available HIF‐1, CD1dhi CD5+ B cells become defective in IL‐10 production; hence, they provide an impaired protection against collagen‐induced arthritis and EAE. HIF‐1 also induces the expression of glycolytic genes, which facilitates the metabolic shift from oxidative phosphorylation to glycolysis, a critical factor for activation and proliferation of CD1dhi CD5+ B cells [72]. Therefore, the function of HIF‐1 in B cells seems to be suppressive in the context of autoimmune pathogenesis (Figure 3).
FIGURE 3.

HIF‐1 modulates the activation of myeloid and B10 cells and therefore contributes to the pro/anti‐inflammatory proportion of Teff versus Treg cells. In M1 macrophages, HIF‐1 induces the expression of pro‐inflammatory cytokines and iNOS. Pro‐inflammatory cytokines induce the differentiation of Teff cells. NO, produced by iNOS, induces the differentiation Treg cells but inhibits the differentiations and proliferation of Teff cells. In dendritic cells, HIF‐1 induces the expression of IL‐10 and glycolytic enzymes. IL‐10 induces the development of Treg cells, and glycolytic enzymes induce the activation and maturation of dendritic cells by shifting the metabolic programme from oxidative phosphorylation to glycolysis. In B10 cells, HIF‐1, in co‐operation with STAT3, enhances the expression of IL‐10, which induces the development of Treg cells. In neutrophils, HIF‐1 induces the expression of pro‐inflammatory cytokines and glycolytic enzymes. Pro‐inflammatory cytokines contribute to the formation of an inflammatory microenvironment within the autoimmune lesions. Pro‐inflammatory cytokines also act on the neutrophils in an autocrine mode and provide anti‐apoptotic survival signal through the activation of NF‐κB and MIP‐β. Glycolytic enzymes, induced by HIF‐1, also contribute to the survival signalling. Longer survival of neutrophils contributes to increased tissue damage in autoimmune diseases. B10, IL‐10‐producing regulatory B cells; HIF‐1, hypoxia‐inducible factor‐1; IL‐10, interleukin‐10; iNOS, inducible nitric oxide synthase; MIP‐β, macrophage inflammatory protein‐β; NF‐κB, nuclear factor kappa‐light‐chain‐enhancer of activated B cells; NO¸nitric oxide; STAT3, signal transducer and activator of transcription 3; Teff, effector T cells; Treg, regulatory T cells
MYELOID CELLS
Although lymphocytes play the most prominent role in the pathogenesis of autoimmune diseases, lymphocytes per se cannot account for the pathological course of the disease in its entirety. Rather, myeloid cells also contribute an inevitable role during induction, mediation and resolution of autoimmune pathogenesis. Macrophages, neutrophils and dendritic cells are the most prominent myeloid cells involved in inflammatory processes. Macrophages have a broad immunomodulatory functions and actively participate in the pathogenesis of many autoimmune diseases [73]. In fact, macrophages are among the earliest immune cells that home into inflamed tissues. Macrophages use pattern recognition receptors (PRRs) to recognize tissue damage or abnormalities by a mechanism in which PRR engagement of endogenous danger‐associated molecular patterns (DAMPs) initiates the creation of an inflammatory microenvironment [74]. In a mature microenvironment, macrophages exhibit a spectrum between two polarized phenotypes, which are referred to as pro‐inflammatory (M1) or anti‐inflammatory (M2) macrophages. Depending on the prevailing local stimuli within the microenvironment (e.g. types of PRR and cytokines produced by infiltrating lymphocytes or injured tissues), macrophages become polarized into M1 or M2 subsets. The relative M1/M2 ratio can potentially shape the progression of disease. Once activated by local stimuli, M1 and M2 macrophages release a wide range of immune mediators. M1 macrophages exert pro‐inflammatory cytotoxic effects, whereas M2 macrophages mediate anti‐inflammatory functions, for example wound repair, tissue remodelling and angiogenesis. M1 macrophages produce reactive ROS/RNS, cytokines (tumour necrosis factor‐α (TNF‐α), IL‐1, IL‐6, IL‐12 and IL‐23) and chemokines (CCL‐5, CXCL9, CXCL10 and CXCL5). Conversely, M2 macrophages produce vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), transforming growth factor‐β (TGF‐β), IL‐4, IL‐13 and IL‐10. This complex milieu of different cytokines and chemokines, produced by macrophages, lays the foundation for the formation of an inflammatory lesion and induces the influx and activation of infiltrating lymphocytes and other myeloid cells [73, 74, 75].
Hypoxia is not a key feature of the inflammatory lesions at the initial stage of their formation. Rather, hypoxia becomes more prominent during the progression of inflammation as the inflammatory sites become metabolically and immunologically more active by the infiltrating myeloid and lymphoid cells. Therefore, the role of HIF‐1 in myeloid cells comes to the prominence at the mature stage of the inflammatory lesions [73, 74, 75]. In contrast to M2 macrophages, HIF‐1 is mainly expressed in M1 macrophages. HIF‐1 induces the expression of pro‐inflammatory cytokines and chemokines in M1 macrophages, which reinforces the pro‐inflammatory nature of the microenvironment [76]. In its mature stage, autoimmune lesions become more populous with pro‐inflammatory infiltrates, which exacerbate the local tissue damage. In addition to cytokines and chemokines, iNOS is one of the most important transcriptional targets of HIF‐1, which is primarily expressed in M1 macrophages [77]. The iNOS enzyme generates nitric oxide (NO) using l‐arginine as substrate. Arginase 1, which is abundantly expressed in M2 macrophages, also uses l‐arginine as substrate to generate ornithine and urea. Expression of arginase 1 is induced by HIF‐2, an isoform of HIF‐1, in M2 macrophages. Because l‐arginine is used as a common substrate by both iNOS and arginase 1, arginase 1 in M2 macrophages indirectly regulates NO production by iNOS in M1 macrophages by limiting the availability of l‐arginine [76]. NO induces the differentiation of NO‐Treg cells (CD4+ CD25+ Foxp3−), which suppresses the proliferation of CD4+CD25− effector T cells in colitis, collagen‐induced arthritis and EAE [78, 79]. In the same time, NO inhibits the differentiation, proliferation and function of Th17 cells through the nitrosylation of aryl hydrocarbon receptor and RORᵞt, and therefore suppresses the diseases in EAE and colitis [54, 80, 81, 82]. Therefore, HIF‐1 in macrophages performs both pro‐inflammatory and immunosuppressive functions in autoimmune lesions (Figure 3). In addition to M1 macrophages, myeloid‐derived suppressor cells (MDSCs) are also well known for their anti‐inflammatory role through the upregulated expression of iNOS. In chronic inflammatory states (e.g. cancer and autoimmune diseases), impaired differentiation of myeloid progenitors into mature myeloid cells leads to the expansion and accumulation of MDSCs [83] MDSCs comprise two subsets including monocytic (M‐MDSCs) and granulocytic/polymorphonuclear (PMN‐MDSCs). Unlike PMN‐MDSCs, M‐MDSCs predominantly express iNOS [83]. Notably, HIF‐1 promotes accumulation of MDSCs in tumour microenvironment, which is hypoxic in nature [84]. Therefore, HIF‐1 may also drive accumulation of MDSCs and therefore play an anti‐inflammatory role in autoimmune lesions; however, additional research is needed to assess this possibility.
Neutrophils are typically the earliest responders to infectious or sterile inflammation (e.g. ischaemia/reperfusion or trauma) [82, 85]. Neutrophils are also known to participate in the pathology of autoimmune diseases of the CNS (e.g. MS, EAE and neuromyelitis optica spectrum disorder). They are prominently present in the autoimmune lesions of the CNS [86, 87, 88, 89, 90]. Neutrophils contribute to chronic inflammatory diseases of CNS by the secretion of pro‐inflammatory cytokines and chemokines, promoting the maturation of antigen‐presenting cells and disruption of the blood–spinal cord barrier [86, 90, 91, 92, 93, 94, 95]. A major regulator of neutrophil function is their short life span through constitutive apoptosis, which is vital for the limitation of tissue damage [96]. Chilvers and colleagues reported that peripheral blood neutrophils become resistant to apoptosis during hypoxia in vitro, which reveals a potential role of HIF‐1 in preserving neutrophil survival and activity in inflammatory environments [97]. Indeed, HIF‐1α‐deficient murine neutrophils displayed a remarkable reduction of cell survival in anoxia, which supported the role of HIF‐1 in the modification of intrinsic apoptotic thresholds in neutrophils. HIF‐1 regulates neutrophil apoptosis by controlling the activation of transcription factor NF‐κB pathway and secretion of macrophage inflammatory protein‐1β, both of which are critically involved in enhancing neutrophil survival, in hypoxic condition through the enhanced expression of an array of extracellular pro‐inflammatory cytokines, for example TNF‐α, IL‐1β and IL‐6 [98, 99, 100]. HIF‐1 also controls neutrophil survival and function through the regulation of metabolic switch to glycolysis, the major ATP‐generating pathway in myeloid cells, through an enhanced expression of glucose transporter 1 and glycolytic enzymes [101]. Taken together, the involvement of neutrophils in the pathology of autoimmune diseases of CNS and the role of HIF‐1 in neutrophil survival and function in response to hypoxia provide evidence that HIF‐1 facilitates the progression of autoimmune diseases by promoting an ameliorated apoptosis and functional persistence of neutrophils (Figure 3).
Myeloid‐derived dendritic cells are centrally involved antigen in presentation and activation of T cells. Tissue‐resident immature dendritic cells become activated and mature upon detection and taking up of antigen patterns in a pro‐inflammatory microenvironment. Mature dendritic cells are characterized by increased antigen presentation, cytokine production, expression of maturation markers and morphologic changes. Upon maturation, dendritic cells migrate to the lymph nodes, and, depending on their surface receptor expression and cytokine profile, they induce the differentiation of subsets of T cell [102, 103]. Under hypoxic condition in inflamed tissue, surface receptor and cytokine profile expressed by dendritic cells may change, which could potentially impact the differentiation of T cells. Hammami et al. [104] showed that HIF‐1α inhibits IL‐12 production in DCs and therefore limits the development of Th1 cells. On the other hand, hypoxia‐activated HIF‐1 in dendritic cells induces the expression of IL‐10 and therefore promotes the differentiation of Treg cells, which is protective in colitis [105]. Like other myeloid cells, the major source of energy generation in mature dendritic cells is glycolysis. In the hypoxic environment of a pro‐inflammatory niche, HIF‐1 in mature dendritic cells mediates a striking increase in glycolysis as compared to basal level, leading to the development of a phenotype, which is optimal for rapid migration and antigen presentation to T cells [106] (Figure 3).
HIF‐1 IS A POTENTIAL THERAPEUTIC TARGET FOR AUTOIMMUNE DISEASES
Critical role of HIF‐1 in immunomodulation of autoimmune diseases has drawn attention towards it as a promising therapeutic target. An emerging literature has revealed the efficacy of HIF‐1‐targeted drugs in inhibiting cancer development, progression and metastasis. HIF‐1‐targeted drugs can be categorized as inhibitors of HIF‐1‐DNA interaction, inhibitors of transcription/translation of HIF‐1 mRNA, inhibitors of HIF‐1‐mediated transcriptional activity or activators of prolyl hydroxylase [107, 108]. Epigallocatechin‐3‐gallate (EGCG) attenuates arthritis in mice by inhibiting STAT3 and HIF‐1α and therefore increasing the relative proportion of Treg over Th17 cells [109]. Under hypoxic condition, NO competes with O2 for binding to mitochondrial cytochrome c oxidase, resulting in the cessation of mitochondrial respiration with the consequence of an increased level of non‐respiratory O2 in the cell. O2 that is not reduced to H2O in oxidative phosphorylation is redistributed and becomes available for prolyl hydroxylation of HIF‐1α leading to its increased proteasomal degradation [110, 111]. Therefore, an exogenous donor of NO such as S‐nitrosoglutathione (GSNO), S‐nitroso‐N‐acetylpenicillamine (SNAP) or diethylenetriamine/nitric oxide adduct (NOC‐18) could be a promising therapeutic option for HIF‐1‐targeted treatment of autoimmune diseases. Antisense oligonucleotide targeting HIF‐1α (EZN‐2698) inhibits tumour growth in xenograft models of human prostate cancer [112]. Topotecan, an FDA‐approved drug currently being used for the chemotherapy of lung cancer and ovarian cancer, inhibits HIF‐1α translation [113]. Echinomycin selectively inhibits the binding of HIF‐1 to DNA showing promise for cancer treatment [114]. Bortezomib (PS‐341), an FDA‐approved drug for treatment of patients with multiple myeloma and patients with mantle cell lymphoma, represses transcriptional activity of HIF‐1α [115]. Although mostly studied for cancer treatment, findings from these studies suggest that HIF‐1 could be a potential target for designing drugs for autoimmune diseases. Therefore, based on the impact of these drugs in cancer, it would be worthwhile to determine whether these inhibitors of HIF‐1 could alter the immune interplay and therefore ameliorate autoimmune diseases in animal models and patients as well. On top of that, considering the divergent roles of HIF‐1 in different immune populations, possibility of potential adverse effects or compromised therapeutic output cannot be ruled out in a HIF‐1‐targeted treatment. Therefore, a lineage‐specific targeting of HIF‐1 might be more effective than the global‐targeting of HIF‐1.
CONCLUSION
Although the exact aetiology and pathogenesis of autoimmune diseases are complex and incompletely understood, a review of the literature reveals abundant evidence that HIF‐1 is pivotal in the modulation of immune mechanisms underlying these diseases. It is well established that inflammation and hypoxia are mutually intertwined. HIF‐1 accumulates under hypoxic condition within the inflammatory lesions and diversely modulates the function of different lymphoid and myeloid cells, which can potentially impact the pathogenesis of autoimmune diseases in a divergent manner. For instance, HIF‐1‐mediated polarization of the balance of Th17/Treg in favour of Treg cells, metabolic switching of lymphocytes towards glycolysis, activation of pro‐inflammatory M1 macrophages, maturation of dendritic cells and reduced apoptosis of neutrophils promote the progression of autoimmune diseases. Conversely, HIF‐1‐induced expansion of CD1dhiCD5+B cells, overexpression of iNOS by M1 macrophages and increased production of IL‐10 by B cells and dendritic cells play anti‐inflammatory roles in autoimmune diseases. Therefore, it deserves further comprehensive investigation to determine how to fine‐tune these contradictory immunomodulatory functions of HIF‐1 in autoimmune diseases by using available therapeutic interventions.
CONFLICT OF INTEREST
The authors have no conflict of interest to declare.
Funding information
IS is supported by the U.S. Department of Veterans Affairs (BX002829, BX003401, and RX002090) and the National Institutes of Health (NS037766).
Contributor Information
S.M. Touhidul Islam, Email: islamt@musc.edu.
Inderjit Singh, Email: singhi@musc.edu.
REFERENCES
- 1.Ziello JE, Jovin IS, Huang Y. Hypoxia‐Inducible Factor (HIF)‐1 regulatory pathway and its potential for therapeutic intervention in malignancy and ischemia. Yale J Biol Med. 2007;80:51–60. [PMC free article] [PubMed] [Google Scholar]
- 2.Wang GL, Jiang BH, Rue EA, Semenza GL. Hypoxia‐inducible factor 1 is a basic‐helix‐loop‐helix‐PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci USA. 1995;92:5510–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Wenger RH, Gassmann M. Oxygen(es) and the hypoxia‐inducible factor‐1. Biol Chem. 1997;378:609–16. [PubMed] [Google Scholar]
- 4.Semenza GL. Hypoxia‐inducible factor 1: oxygen homeostasis and disease pathophysiology. Trends Mol Med. 2001;7:345–50. [DOI] [PubMed] [Google Scholar]
- 5.Kominsky DJ, Campbell EL, Colgan SP. Metabolic shifts in immunity and inflammation. J Immunol. 2010;184:4062–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bartels K, Grenz A, Eltzschig HK. Hypoxia and inflammation are two sides of the same coin. Proc Natl Acad Sci USA. 2013;110:18351–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Eltzschig HK, Carmeliet P. Hypoxia and inflammation. N Engl J Med. 2011;364:656–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Fox CJ, Hammerman PS, Thompson CB. Fuel feeds function: energy metabolism and the T‐cell response. Nat Rev Immunol. 2005;5:844–52. [DOI] [PubMed] [Google Scholar]
- 9.Pollard TD, Borisy GG. Cellular motility driven by assembly and disassembly of actin filaments. Cell. 2003;112:453–65. [DOI] [PubMed] [Google Scholar]
- 10.Borregaard N, Herlin T. Energy metabolism of human neutrophils during phagocytosis. J Clin Invest. 1982;70:550–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Robinson JM. Reactive oxygen species in phagocytic leukocytes. Histochem Cell Biol. 2008;130:281–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Greiner EF, Guppy M, Brand K. Glucose is essential for proliferation and the glycolytic enzyme induction that provokes a transition to glycolytic energy production. J Biol Chem. 1994;269:31484–90. [PubMed] [Google Scholar]
- 13.Forstermann U, Sessa WC. Nitric oxide synthases: regulation and function. Eur Heart J. 2012;33:pp. 829–37, 37a–37d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Gabig TG, Bearman SI, Babior BM. Effects of oxygen tension and pH on the respiratory burst of human neutrophils. Blood. 1979;53:1133–9. [PubMed] [Google Scholar]
- 15.Xu Q, Choksi S, Qu J, Jang J, Choe M, Banfi B, et al. NADPH oxidases are essential for macrophage differentiation. J Biol Chem. 2016;291:20030–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Belambri SA, Rolas L, Raad H, Hurtado‐Nedelec M, Dang PM, El‐Benna J. NADPH oxidase activation in neutrophils: Role of the phosphorylation of its subunits. Eur J Clin Invest. 2018;48(Suppl 2):e12951. [DOI] [PubMed] [Google Scholar]
- 17.Salvemini D, Doyle TM, Cuzzocrea S. Superoxide, peroxynitrite and oxidative/nitrative stress in inflammation. Biochem Soc Trans. 2006;34:965–70. [DOI] [PubMed] [Google Scholar]
- 18.Pacher P, Beckman JS, Liaudet L. Nitric oxide and peroxynitrite in health and disease. Physiol Rev. 2007;87:315–424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Marshall JS, Warrington R, Watson W, Kim HL. An introduction to immunology and immunopathology. Allergy Asthma Clin Immunol. 2018;14:49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Deng W, Feng X, Li X, Wang D, Sun L. Hypoxia‐inducible factor 1 in autoimmune diseases. Cell Immunol. 2016;303:7–15. [DOI] [PubMed] [Google Scholar]
- 21.Quiñonez‐Flores CM, González‐Chávez SA, Pacheco‐Tena C. Hypoxia and its implications in rheumatoid arthritis. J Biomed Sci. 2016;23:62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wekerle H. Tissue hypoxia in autoimmune brain disease: a Columbian discovery? Ann Neurol. 2013;74:765–7. [DOI] [PubMed] [Google Scholar]
- 23.Konisti S, Kiriakidis S, Paleolog EM. Hypoxia–a key regulator of angiogenesis and inflammation in rheumatoid arthritis. Nat Rev Rheumatol. 2012;8:153–62. [DOI] [PubMed] [Google Scholar]
- 24.Karhausen J, Furuta GT, Tomaszewski JE, Johnson RS, Colgan SP, Haase VH. Epithelial hypoxia‐inducible factor‐1 is protective in murine experimental colitis. Journal of Clinical Investigation. 2004;114:1098–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Rosenberger C, Solovan C, Rosenberger AD, Jinping L, Treudler R, Frei U, et al. Upregulation of hypoxia‐inducible factors in normal and psoriatic skin. J Invest Dermatol. 2007;127:2445–52. [DOI] [PubMed] [Google Scholar]
- 26.Distler O, Distler JHW, Scheid A, Acker T, Hirth A, Rethage J, et al. Uncontrolled expression of vascular endothelial growth factor and its receptors leads to insufficient skin angiogenesis in patients with systemic sclerosis. Circ Res. 2004;95:109–16. [DOI] [PubMed] [Google Scholar]
- 27.Yao S‐Y, Soutto M, Sriram S. Preconditioning with cobalt chloride or desferrioxamine protects oligodendrocyte cell line (MO3.13) from tumor necrosis factor‐α‐mediated cell death. J Neurosci Res. 2008;86:2403–13. [DOI] [PubMed] [Google Scholar]
- 28.Loukovaara S, Koivunen P, Ingles M, Escobar J, Vento M, Andersson S. Elevated protein carbonyl and HIF‐1alpha levels in eyes with proliferative diabetic retinopathy. Acta Ophthalmol. 2014;92:323–7. [DOI] [PubMed] [Google Scholar]
- 29.Muz B, Khan MN, Kiriakidis S, Hypoxia PEM. The role of hypoxia and HIF‐dependent signalling events in rheumatoid arthritis. Arthritis Res Ther. 2009;11:201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Giatromanolaki A, Sivridis E, Maltezos E, Papazoglou D, Simopoulos C, Gatter KC, et al. Hypoxia inducible factor 1α and 2α overexpression in inflammatory bowel disease. J Clin Pathol. 2003;56:209–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Ioannou M, Pyrpasopoulou A, Simos G, Paraskeva E, Nikolaidou C, Venizelos I, et al. Upregulation of VEGF expression is associated with accumulation of HIF‐1alpha in the skin of naive scleroderma patients. Mod Rheumatol. 2013;23:1245–8. [DOI] [PubMed] [Google Scholar]
- 32.Graumann U, Reynolds R, Steck AJ, Schaeren‐Wiemers N. Molecular changes in normal appearing white matter in multiple sclerosis are characteristic of neuroprotective mechanisms against hypoxic insult. Brain Pathol. 2003;13:554–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Liu W, Shen SM, Zhao XY, Chen GQ. Targeted genes and interacting proteins of hypoxia inducible factor‐1. Int J Biochem Mol Biol. 2012;3:165–78. [PMC free article] [PubMed] [Google Scholar]
- 34.Greijer AE, van der Wall E. The role of hypoxia inducible factor 1 (HIF‐1) in hypoxia induced apoptosis. J Clin Pathol. 2004;57:1009–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Hellwig‐Burgel T, Stiehl DP, Wagner AE, Metzen E, Jelkmann W. Review: hypoxia‐inducible factor‐1 (HIF‐1): a novel transcription factor in immune reactions. J Interferon Cytokine Res. 2005;25:297–310. [DOI] [PubMed] [Google Scholar]
- 36.Dardalhon V, Korn T, Kuchroo VK, Anderson AC. Role of Th1 and Th17 cells in organ‐specific autoimmunity. J Autoimmun. 2008;31:252–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Fife BT, Pauken KE, Eagar TN, Obu T, Wu J, Tang Q, et al. Interactions between PD‐1 and PD‐L1 promote tolerance by blocking the TCR‐induced stop signal. Nat Immunol. 2009;10:1185–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Weaver CT, Hatton RD, Mangan PR, Harrington LE. IL‐17 family cytokines and the expanding diversity of effector T cell lineages. Annu Rev Immunol. 2007;25:821–52. [DOI] [PubMed] [Google Scholar]
- 39.Zhuang Z, Wang Y, Zhu G, Gu Y, Mao L, Hong M, et al. Imbalance of Th17/Treg cells in pathogenesis of patients with human leukocyte antigen B27 associated acute anterior uveitis. Sci Rep. 2017;7:40414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Barnes MJ, Powrie F. Regulatory T cells reinforce intestinal homeostasis. Immunity. 2009;31:401–11. [DOI] [PubMed] [Google Scholar]
- 41.Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell. 2008;133:775–87. [DOI] [PubMed] [Google Scholar]
- 42.Ahern PP, Schiering C, Buonocore S, McGeachy MJ, Cua DJ, Maloy KJ, et al. Interleukin‐23 drives intestinal inflammation through direct activity on T cells. Immunity. 2010;33:279–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, et al. Conversion of peripheral CD4+CD25‐ naive T cells to CD4+CD25+ regulatory T cells by TGF‐beta induction of transcription factor Foxp3. J Exp Med. 2003;198:1875–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Luo X, Zhang Q, Liu V, Xia Z, Pothoven KL, Lee C. Cutting edge: TGF‐beta‐induced expression of Foxp3 in T cells is mediated through inactivation of ERK. J Immunol. 2008;180:2757–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Rajewsky K, von Boehmer H. Lymphocyte development: overview. Curr Opin Immunol. 2008;20:127–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Dang Eric V, Barbi J, Yang H‐Y, Jinasena D, Yu H, Zheng Y, et al. Control of TH17/Treg balance by hypoxia‐inducible factor 1. Cell. 2011;146:772–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zhou L, Lopes JE, Chong MM, Ivanov II, Min R, Victora GD, et al. TGF‐beta‐induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function. Nature. 2008;453:236–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGFβ in the context of an inflammatory cytokine milieu supports de novo differentiation of IL‐17‐producing T cells. Immunity. 2006;24:179–89. [DOI] [PubMed] [Google Scholar]
- 49.Yang XO, Nurieva R, Martinez GJ, Kang HS, Chung Y, Pappu BP, et al. Molecular antagonism and plasticity of regulatory and inflammatory T cell programs. Immunity. 2008;29:44–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Ichiyama K, Yoshida H, Wakabayashi Y, Chinen T, Saeki K, Nakaya M, et al. Foxp3 inhibits RORgammat‐mediated IL‐17A mRNA transcription through direct interaction with RORgammat. J Biol Chem. 2008;283:17003–8. [DOI] [PubMed] [Google Scholar]
- 51.Jung JE, Lee HG, Cho IH, Chung DH, Yoon SH, Yang YM, et al. STAT3 is a potential modulator of HIF‐1‐mediated VEGF expression in human renal carcinoma cells. FASEB J. 2005;19:1296–8. [DOI] [PubMed] [Google Scholar]
- 52.Shi LZ, Wang R, Huang G, Vogel P, Neale G, Green DR, et al. HIF1α–dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J Exp Med. 2011;208:1367–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Cluxton D, Petrasca A, Moran B, Fletcher JM. Differential regulation of human Treg and Th17 Cells by fatty acid synthesis and glycolysis. Front Immunol. 2019;10:115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Jianjun Y, Zhang R, Lu G, Shen Y, Peng L, Zhu C, et al. T cell‐derived inducible nitric oxide synthase switches off Th17 cell differentiation. J Exp Med. 2013;210:1447–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Wenger RH. Cellular adaptation to hypoxia: O2‐sensing protein hydroxylases, hypoxia‐inducible transcription factors, and O2‐regulated gene expression. FASEB J. 2002;16:1151–62. [DOI] [PubMed] [Google Scholar]
- 56.Clambey ET, McNamee EN, Westrich JA, Glover LE, Campbell EL, Jedlicka P, et al. Hypoxia‐inducible factor‐1 alpha–dependent induction of FoxP3 drives regulatory T‐cell abundance and function during inflammatory hypoxia of the mucosa. Proc Natl Acad Sci USA. 2012;109:E2784–E2793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Damsker JM, Hansen AM, Caspi RR. Th1 and Th17 cells: adversaries and collaborators. Ann N Y Acad Sci. 2010;1183:211–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Shehade H, Acolty V, Moser M, Oldenhove G. Cutting edge: hypoxia‐inducible Factor 1 negatively regulates Th1 function. J Immunol. 2015;195:1372–6. [DOI] [PubMed] [Google Scholar]
- 59.Yang C, Mai H, Peng J, Zhou B, Hou J, Jiang D. STAT4: an immunoregulator contributing to diverse human diseases. Int J Biol Sci. 2020;16:1575–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Bharadwaj U, Kasembeli MM, Robinson P, Tweardy DJ. Targeting Janus Kinases and signal transducer and activator of transcription 3 to treat inflammation, fibrosis, and cancer: rationale, progress, and caution. Pharmacol Rev. 2020;72:486–526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Thieu VT, Yu Q, Chang HC, Yeh N, Nguyen ET, Sehra S, et al. Signal transducer and activator of transcription 4 is required for the transcription factor T‐bet to promote T helper 1 cell‐fate determination. Immunity. 2008;29:679–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Roncarolo MG, Gregori S, Battaglia M, Bacchetta R, Fleischhauer K, Levings MK. Interleukin‐10‐secreting type 1 regulatory T cells in rodents and humans. Immunol Rev. 2006;212:28–50. [DOI] [PubMed] [Google Scholar]
- 63.Jia X, Zhai T, Wang B, Yao Q, Li Q, Mu K, et al. Decreased number and impaired function of type 1 regulatory T cells in autoimmune diseases. J Cell Physiol. 2019;234:12442–50. [DOI] [PubMed] [Google Scholar]
- 64.Meka RR, Venkatesha SH, Dudics S, Acharya B, Moudgil KD. IL‐27‐induced modulation of autoimmunity and its therapeutic potential. Autoimmun Rev. 2015;14:1131–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Wang H, Meng R, Li Z, Yang B, Liu Y, Huang F, et al. IL‐27 induces the differentiation of Tr1‐like cells from human naive CD4+ T cells via the phosphorylation of STAT1 and STAT3. Immunol Lett. 2011;136:21–8. [DOI] [PubMed] [Google Scholar]
- 66.Mascanfroni ID, Takenaka MC, Yeste A, Patel B, Wu Y, Kenison JE, et al. Metabolic control of type 1 regulatory T cell differentiation by AHR and HIF1‐alpha. Nat Med. 2015;21:638–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Apetoh L, Quintana FJ, Pot C, Joller N, Xiao S, Kumar D, et al. The aryl hydrocarbon receptor interacts with c‐Maf to promote the differentiation of type 1 regulatory T cells induced by IL‐27. Nat Immunol. 2010;11:854–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Pot C, Apetoh L, Kuchroo VK. Type 1 regulatory T cells (Tr1) in autoimmunity. Semin Immunol. 2011;23:202–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Atarashi K, Nishimura J, Shima T, Umesaki Y, Yamamoto M, Onoue M, et al. ATP drives lamina propria T(H)17 cell differentiation. Nature. 2008;455:808–12. [DOI] [PubMed] [Google Scholar]
- 70.Dang EV, Barbi J, Yang HY, Jinasena D, Yu H, Zheng Y, et al. Control of T(H)17/T(reg) balance by hypoxia‐inducible factor 1. Cell. 2011;146:772–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Doedens AL, Phan AT, Stradner MH, Fujimoto JK, Nguyen JV, Yang E, et al. Hypoxia‐inducible factors enhance the effector responses of CD8(+) T cells to persistent antigen. Nat Immunol. 2013;14:1173–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Meng X, Grotsch B, Luo Y, Knaup KX, Wiesener MS, Chen XX, et al. Hypoxia‐inducible factor‐1alpha is a critical transcription factor for IL‐10‐producing B cells in autoimmune disease. Nat Commun. 2018;9:251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ma WT, Gao F, Gu K, Chen DK. The role of monocytes and macrophages in autoimmune diseases: a comprehensive review. Front Immunol. 2019;10:1140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Funes SC, Rios M, Escobar‐Vera J, Kalergis AM. Implications of macrophage polarization in autoimmunity. Immunology. 2018;154:186–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Navegantes KC, de Souza GR, Pereira PAT, Czaikoski PG, Azevedo CHM, Monteiro MC. Immune modulation of some autoimmune diseases: the critical role of macrophages and neutrophils in the innate and adaptive immunity. J Transl Med. 2017;15:36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Takeda N, O'Dea EL, Doedens A, Kim JW, Weidemann A, Stockmann C, et al. Differential activation and antagonistic function of HIF‐{alpha} isoforms in macrophages are essential for NO homeostasis. Genes Dev. 2010;24:491–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Gordon S. Alternative activation of macrophages. Nat Rev Immunol. 2003;3:23–35. [DOI] [PubMed] [Google Scholar]
- 78.Niedbala W, Cai B, Liu H, Pitman N, Chang L, Liew FY. Nitric oxide induces CD4+CD25+ Foxp3 regulatory T cells from CD4+CD25 T cells via p53, IL‐2, and OX40. Proc Natl Acad Sci USA. 2007;104:15478–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Niedbala W, Besnard AG, Jiang HR, Alves‐Filho JC, Fukada SY, Nascimento D, et al. Nitric oxide‐induced regulatory T cells inhibit Th17 but not Th1 cell differentiation and function. J Immunol. 2013;191:164–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Nath N, Morinaga O, Singh I. S‐nitrosoglutathione a physiologic nitric oxide carrier attenuates experimental autoimmune encephalomyelitis. J Neuroimmune Pharmacol. 2010;5:240–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Niedbala W, Alves‐Filho JC, Fukada SY, Vieira SM, Mitani A, Sonego F, et al. Regulation of type 17 helper T‐cell function by nitric oxide during inflammation. Proc Natl Acad Sci USA. 2011;108:9220–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Singh I, Nath N, Saxena N, Singh AK, Won JS. Regulation of IL‐10 and IL‐17 mediated experimental autoimmune encephalomyelitis by S‐nitrosoglutathione. Immunobiology. 2018;223:549–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Consonni FM, Porta C, Marino A, Pandolfo C, Mola S, Bleve A, et al. Myeloid‐derived suppressor cells: ductile targets in disease. Front Immunol. 2019;10:949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Chiu DK‐C, Tse AP‐W, Xu IM‐J, Di Cui J, Lai RK‐H, Li LL, et al. Hypoxia inducible factor HIF‐1 promotes myeloid‐derived suppressor cells accumulation through ENTPD2/CD39L1 in hepatocellular carcinoma. Nature. Communications. 2017;8:517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Wang J, Hossain M, Thanabalasuriar A, Gunzer M, Meininger C, Kubes P. Visualizing the function and fate of neutrophils in sterile injury and repair. Science. 2017;358:111–6. [DOI] [PubMed] [Google Scholar]
- 86.Hertwig L, Pache F, Romero‐Suarez S, Stürner KH, Borisow N, Behrens J, et al. Distinct functionality of neutrophils in multiple sclerosis and neuromyelitis optica. Mult Scler J. 2016;22:160–73. [DOI] [PubMed] [Google Scholar]
- 87.Pierson ER, Wagner CA, Goverman JM. The contribution of neutrophils to CNS autoimmunity. Clin Immunol. 2018;189:23–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Saadoun S, Waters P, MacDonald C, Bell BA, Vincent A, Verkman AS, et al. Neutrophil protease inhibition reduces neuromyelitis optica–immunoglobulin G–induced damage in mouse brain. Ann Neurol. 2012;71:323–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Miller NM, Wang J, Tan Y, Dittel BN. Anti‐inflammatory mechanisms of IFN‐gamma studied in experimental autoimmune encephalomyelitis reveal neutrophils as a potential target in multiple sclerosis. Front Neurosci. 2015;9:287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Yan Z, Yang W, Parkitny L, Gibson SA, Lee KS, Collins F, et al. Deficiency of Socs3 leads to brain‐targeted EAE via enhanced neutrophil activation and ROS production. JCI Insight. 2019;5:e126520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Lévesque SA, Paré A, Mailhot B, Bellver‐Landete V, Kébir H, Lécuyer M‐A, et al. Myeloid cell transmigration across the CNS vasculature triggers IL‐1β–driven neuroinflammation during autoimmune encephalomyelitis in mice. J Exp Med. 2016;213:929–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Jiang W, St‐Pierre S, Roy P, Morley BJ, Hao J, Simard AR. Infiltration of CCR2+Ly6Chigh proinflammatory monocytes and neutrophils into the central nervous system is modulated by nicotinic acetylcholine receptors in a model of multiple sclerosis. J Immunol. 2016;196:2095–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Steinbach K, Piedavent M, Bauer S, Neumann JT, Friese MA. Neutrophils amplify autoimmune central nervous system infiltrates by maturing local APCs. J Immunol. 2013;191:4531–9. [DOI] [PubMed] [Google Scholar]
- 94.Aubé B, Lévesque SA, Paré A, Chamma É, Kébir H, Gorina R, et al. Neutrophils mediate blood‐spinal cord barrier disruption in demyelinating neuroinflammatory diseases. J Immunol. 2014;193:2438–54. [DOI] [PubMed] [Google Scholar]
- 95.Rumble JM, Huber AK, Krishnamoorthy G, Srinivasan A, Giles DA, Zhang X, et al. Neutrophil‐related factors as biomarkers in EAE and MS. J Exp Med. 2015;212:23–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Haslett C. Resolution of acute inflammation and the role of apoptosis in the tissue fate of granulocytes. Clin Sci (Lond). 1992;83:639–48. [DOI] [PubMed] [Google Scholar]
- 97.Mecklenburgh KI, Walmsley SR, Cowburn AS, Wiesener M, Reed BJ, Upton PD, et al. Involvement of a ferroprotein sensor in hypoxia‐mediated inhibition of neutrophil apoptosis. Blood. 2002;100:3008–16. [DOI] [PubMed] [Google Scholar]
- 98.Ward C, Chilvers ER, Lawson MF, Pryde JG, Fujihara S, Farrow SN, et al. NF‐kappaB activation is a critical regulator of human granulocyte apoptosis in vitro. J Biol Chem. 1999;274:4309–18. [DOI] [PubMed] [Google Scholar]
- 99.Walmsley SR, Print C, Farahi N, Peyssonnaux C, Johnson RS, Cramer T, et al. Hypoxia‐induced neutrophil survival is mediated by HIF‐1alpha‐dependent NF‐kappaB activity. J Exp Med. 2005;201:105–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.D'Ignazio L, Bandarra D, Rocha S. NF‐kappaB and HIF crosstalk in immune responses. FEBS J. 2016;283:413–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Cramer T, Yamanishi Y, Clausen BE, Forster I, Pawlinski R, Mackman N, et al. HIF‐1alpha is essential for myeloid cell‐mediated inflammation. Cell. 2003;112:645–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Morel PA, Butterfield LH. Dendritic cell control of immune responses. Front Immunol. 2015;6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Mildner A, Jung S. Development and function of dendritic cell subsets. Immunity. 2014;40:642–56. [DOI] [PubMed] [Google Scholar]
- 104.Hammami A, Abidin BM, Heinonen KM, Stager S. HIF‐1alpha hampers dendritic cell function and Th1 generation during chronic visceral leishmaniasis. Sci Rep. 2018;8:3500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Flück K, Breves G, Fandrey J, Winning S. Hypoxia‐inducible factor 1 in dendritic cells is crucial for the activation of protective regulatory T cells in murine colitis. Mucosal Immunol. 2016;9:379–90. [DOI] [PubMed] [Google Scholar]
- 106.Stothers CL, Luan L, Fensterheim BA, Bohannon JK. Hypoxia‐inducible factor‐1alpha regulation of myeloid cells. J Mol Med (Berl). 2018;96:1293–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Xia Y, Choi H‐K, Lee K. Recent advances in hypoxia‐inducible factor (HIF)‐1 inhibitors. Eur J Med Chem. 2012;49:24–40. [DOI] [PubMed] [Google Scholar]
- 108.Onnis B, Rapisarda A, Melillo G. Development of HIF‐1 inhibitors for cancer therapy. J Cell Mol Med. 2009;13(9A):2780–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Yang EJ, Lee J, Lee SY, Kim EK, Moon YM, Jung YO, et al. EGCG attenuates autoimmune arthritis by inhibition of STAT3 and HIF‐1alpha with Th17/Treg control. PLoS One. 2014;9:e86062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Hagen T, Taylor CT, Lam F, Moncada S. Redistribution of intracellular oxygen in hypoxia by nitric oxide: effect on HIF1alpha. Science. 2003;302:1975–8. [DOI] [PubMed] [Google Scholar]
- 111.Callapina M, Zhou J, Schmid T, Kohl R, Brune B. NO restores HIF‐1alpha hydroxylation during hypoxia: role of reactive oxygen species. Free Radic Biol Med. 2005;39:925–36. [DOI] [PubMed] [Google Scholar]
- 112.Greenberger LM, Horak ID, Filpula D, Sapra P, Westergaard M, Frydenlund HF, et al. A RNA antagonist of hypoxia‐inducible factor‐1alpha, EZN‐2968, inhibits tumor cell growth. Mol Cancer Ther. 2008;7:3598–608. [DOI] [PubMed] [Google Scholar]
- 113.Rapisarda A, Uranchimeg B, Scudiero DA, Selby M, Sausville EA, Shoemaker RH, et al. Identification of small molecule inhibitors of hypoxia‐inducible factor 1 transcriptional activation pathway. Cancer Res. 2002;62:4316–24. [PubMed] [Google Scholar]
- 114.Kong D, Park EJ, Stephen AG, Calvani M, Cardellina JH, Monks A, et al. Echinomycin, a small‐molecule inhibitor of hypoxia‐inducible factor‐1 DNA‐binding activity. Cancer Res. 2005;65:9047–55. [DOI] [PubMed] [Google Scholar]
- 115.Shin DH, Chun YS, Lee DS, Huang LE, Park JW. Bortezomib inhibits tumor adaptation to hypoxia by stimulating the FIH‐mediated repression of hypoxia‐inducible factor‐1. Blood. 2008;111:3131–6. [DOI] [PubMed] [Google Scholar]
