Skip to main content
Experimental Biology and Medicine logoLink to Experimental Biology and Medicine
. 2022 Sep 23;247(24):2184–2191. doi: 10.1177/15353702221121635

The expression and function of growth hormone secretagogue receptor in immune cells: A current perspective

Ji Yeon Noh 1, Matthew Herrera 1, Bhimanagouda S Patil 2, Xiao-Di Tan 3, Gus A Wright 4, Yuxiang Sun 1,5,
PMCID: PMC9899990  PMID: 36151745

Abstract

The orexigenic hormone ghrelin and its receptor, growth hormone secretagogue receptor (GHS-R), have been extensively studied in the last two decades, revealing that ghrelin signaling has important implications in health and disease. Metabolic diseases, such as obesity and diabetes, are often accompanied by low-grade chronic inflammation, that has been coined as “meta-inflammation.” Immune cells are key cellular mediators of meta-inflammation, controlling both initiation and resolution of inflammation. Immune cells exhibit dynamic changes in cellular characteristics and functional output in response to the stimuli/insults from their surrounding microenvironment. Emerging evidence shows that ghrelin has an important effect on inflammation, in addition to its well-known effects on metabolism. However, the cellular/molecular mechanism of ghrelin signaling in immunity is largely unknown because the knowledge in regard to the expression and function of GHS-R in immune cells is currently sparse. In this review, we have accumulated the recent findings related to the expression and functions of GHS-R in various immune cells under different physiological and pathological states. This review aims to inspire further investigation of the immunological roles of ghrelin signaling and advance the therapeutic applications of ghrelin signaling in meta-inflammation.

Keywords: Ghrelin, growth hormone secretagogue receptor (GHS-R), immune cells, immunometabolism, inflammaging, polarization, meta-inflammation, macrophages, neutrophils, dendritic cells, T lymphocytes, B lymphocytes

Impact Statement

Emerging evidence shows that ghrelin and growth hormone secretagogue receptor (GHS-R) play significant roles in inflammation. Ghrelin in the circulation can reach all parts of the body, and it signals through its receptor GHS-R in tissues. To understand the role and mechanism of ghrelin signaling in inflammation, it is critical to identify the immune cells that express GHS-R, since the receptor expression predicts the site of action and specifies the effects. However, the current data on GHS-R expression in immune cells are sparse and confounding. In this review, we summarize the emerging findings in the literature regarding GHS-R expression in common immune cells, and discuss the current understanding of the effects of GHS-R in immune cells under different states. We believe that our perspective in this review will help to guide the future research of ghrelin signaling in immunity, thus to advance the understanding and application of ghrelin signaling in inflammatory diseases.

Introduction

Ghrelin, aka orexigenic hormone or hunger hormone, classically is known for its roles in the regulation of appetite and adiposity. 1 Ghrelin is extensively studied by our group and others as an important regulator of glucose homeostasis 2 and energy homeostasis.110 As we previously reviewed that ghrelin is a multifaced hormone and ghrelin signaling has a broad range of functions. 11 In addition to regulating metabolism, ghrelin signaling also regulates cardiac function, 12 muscle mass, 13 bone density, 14 and cancer. 15 Ghrelin gene produces three protein isoforms by alternative splicing or post-translational modification: acyl-ghrelin (aka active ghrelin, ghrelin), unacyl ghrelin (UAG, aka inactive ghrelin), and obestatin.11,16,17 Acyl-ghrelin is active form of ghrelin, often simply referred as ghrelin; it binds specifically to growth hormone secretagogue receptor (GHS-R),1,18 and acylation of ghrelin is required for its binding to GHS-R.19,20 UAG is considered as inactive form due to low binding affinity to GHS-R. 21 The functional receptor for obestatin is still an ongoing debate, as multiple receptors have been proposed including GHS-R, 22 glucagon-like peptide 1 receptor, 23 and G-protein coupled receptor 39. 24 While ghrelin function as agonist for GHS-R, liver-expressed antimicrobial peptide 2 (LEAP-2) has now been identified as antagonist for GHS-R. 25 GHS-R has two isomers: GHS-R1a and GHS-R1b; GHS-R1a is considered as the functionally relevant receptor because it binds to ghrelin, whereas GHS-R1b does not.26,27 It has been shown that GHS-R1b has an inhibitory effect on GHS-R1a signaling and can form heterodimeric complexes with other receptors. 28 In this current review, we focus on acyl-ghrelin (referring it as ghrelin), because it is the only confirmed endogenous agonist for GHS-R, and limit our discussion on the functionally relevant receptor GHS-R1a (referring as GHS-R).

Immune cells have critical roles in normal and disease conditions; in this review, we primarily focus on the role of ghrelin signaling in chronic inflammation, aka meta-inflammation, that is prevalent to obesity-associated diseases and aging.2932 Immune cells – such as macrophages, dendritic cells (DCs), neutrophils, T cells, and B cells – have been shown to play critical roles in the initiation and resolution of inflammation under diet-induced obesity (DIO).3336 In obesity, the infiltration of macrophages and DCs into adipose tissues is increased exponentially,30,37,38 and macrophages shift toward a pro-inflammatory state and secrete pro-inflammatory cytokines. 30 In aging, immune dysfunction in the elderly is evident, showing reduced responsiveness to vaccination, decreased immune surveillance, reduced resistance to infections, but increased susceptibility to inflammation and autoimmune activation.31,39 Another hallmark impairment of the immune system in aging is thymic atrophy. Thymic atrophy causes increased output of self-reactive T cells, reduced autoimmune suppression by regulatory T cells, and decreased naïve T cells; these pathological changes exacerbate inflammation and immunosenescence, promoting inflammaging. 32 Thus, immune cells have critical roles in the pathogenesis of diseases and aging. It is extremely important to understand the expression and roles of GHS-R in immune cells in order to further elucidate the immune regulation of ghrelin signaling in inflammatory diseases and aging.

Ghrelin and GHS-R expressing tissues and cells

Ghrelin is highly produced by X/A-like cells, a group of unique endocrine cells in gastric mucosa of the stomach, and it is also ubiquitously expressed throughout the body.40,41 In contrast, GHS-R expression is much more restricted. In mice, the highest expression of GHS-R is detected in the pituitary gland and the hypothalamus,4244 and low levels of GHS-R expression are detected in selective peripheral tissues, such as heart, thymus, testes, lung, adrenal, small intestine, spleen, and kidney.43,45 We reported that GHS-R is detected in mouse bone marrow, pancreas, skeletal muscle, brown adipose tissue, and white adipose tissue, whereas GHS-R expression level in those peripheral tissues is much lower compared to that in pituitary and brain. 42 The tissues expressing ghrelin and GHS-R in humans and rodents are, respectively, summarized in Table 1. While ghrelin, as a hormone, can reach all tissues through circulation, GHS-R is a cell-surface receptor that is expressed in specific cell types to mediate down-stream signaling events. It needs to be emphasized that the data of GHS-R expression are more limited compared to that of ghrelin, and the GHS-R expression data in Table 1 do not pertain to specific individual cell types, but rather to the whole tissue that includes different types of cells (including various immune cells). For example, in liver, while the predominant cell type is hepatocytes, it also contains immune cells, such as Kupffer cells, innate lymphoid cells (ILCs), DCs, natural killer (NK) cells, invariant NKT (iNKT) cells, mucosal-associated invariant T (MAIT) cells, and γδ T cells.46,47 Therefore, caution need to be taken when interpreting the GHS-R expression data in tissue.

Table 1.

Ghrelin and GHS-R expressing tissues in human, mouse, and rat.

Expression Species Tissue types Detection method References
Ghrelin Human Stomach, duodenum, jejunum, ileum, colon, esophagus, buccal, antrum, lung, pancreas, lymph node, spleen, liver, gallbladder, pituitary, somatotropinoma, breast, kidney, adrenal, ovary, fallopian tubes, testis, prostate, adipose tissue, placenta, muscle, bladder, thyroid, thymus, atrium, myocardium, skin RT-PCR 40,41
Mouse/rat Stomach, duodenum, jejunum, ileum, cecum, colon, submaxillary gland, lung, pancreas, liver, hypothalamus, pituitary, kidney, adrenal, ovary, testis, thyroid, adipose tissue, muscle, thymus, atrium, ventricle, aorta, plasma RT-PCR
RIA
16,43–45,77
GHS-R Human Pituitary, hypothalamus, hippocampal formation, cerebral cortex, basal ganglia, small intestine, rectum, appendix, lung, pancreas, spleen, gallbladder, adrenal, bone marrow, testis, skin RT-PCR 79
Mouse/rat Pituitary, hypothalamus, brain, small intestine, spleen, lung, pancreas, kidney, adrenal, heart, thymus, bone marrow, skeletal muscle, brown adipose tissue, white adipose tissue, testes RT-PCR 42–44,77

GHS-R: growth hormone secretagogue receptor; RT-PCR: reverse transcription polymerase chain reaction; RIA: radioimmunoassay.

It is important to note that the GHS-R expression above does not pertain to specific cell types, but rather reflects the levels in the whole tissue that includes different types of cells, including immune cells.

Expression of ghrelin and GHS-R under chronic inflammatory states

The notion that ghrelin signaling is relevant to metabolic diseases and aging is based on the evidences reported by our group and others on the tissue expression profile of ghrelin and GHS-R under obesity and aging, and the identified functions with loss or gain of function studies of ghrelin/GHS-R.1,3,9,43,4853 We showed that mouse plasma ghrelin levels are increased with aging.43,48 Counterintuitively, plasma ghrelin levels are reduced under DIO,51,52 and the ghrelin-producing cells in the stomachs of mice are decreased under DIO. 53 Consistent with the mouse data, circulating ghrelin levels in obese humans are also reduced. 50 A recent report uncovered that decreased ghrelin levels in obesity are due to the fact that the insulin receptor in ghrelin-expressing cells inhibits ghrelin secretion under obesity. 54

We showed that GHS-R gene expression in mouse pituitary glands is highly variable throughout aging: with the highest expression in one to two months old mice, an increasing trend with aging between 6 and 28 months of age, while the expression at 28 months of age is still lower than that of one to two months old young mice. 43 In addition, we reported that high-fat diet feeding significantly increased the expression of GHS-R in the arcuate nucleus and ventromedial hypothalamus in the mouse brain. 3 Interestingly, another study reported that the GHS-R mRNA levels in the nodose ganglion and hypothalamus of high-fat diet fed mice were lower compared to that of chow-fed mice. 49 Importantly, we demonstrated that GHS-R ablation protects against DIO, and ameliorates age-associated inflammation and insulin resistance.3,42,55 These results collectively indicate that ghrelin/GHS-R signaling has important roles in diet- and aging-associated meta-inflammation and metabolic dysfunctions.

As mentioned above that hormone ghrelin is produced ubiquitously in almost all tissues, including immune cells, the highest expression of ghrelin is detected in stomach. 41 Regardless of the site of production, as a hormone, ghrelin can reach all target cells through circulation in the body. Ghrelin’s function is mediated by its receptor GHS-R. In contrast to ghrelin, the expression of GHS-R is restricted to specific cell types; the highest expression of GHS-R is in the neurons of brain.42,43 Among other selected cell types expressing GHS-R, immune cells, including macrophages 42 and T cells, 56 have the second highest expression. The site of GHS-R expression dictates where the ligand–receptor interaction takes place. Thus, ghrelin can regulate immune cells by endocrine and/or autocrine mechanism. It is essential to determine the sites and levels of GHS-R expression under physiological/pathological conditions in order to understand the regulatory roles of ghrelin signaling in pathogenesis and disease progression of various diseases, and facilitate the development of potential cell-specific therapeutic interventions. Table 2 summarizes the expression level changes of ghrelin and GHS-R under various chronic inflammatory conditions, such as DIO, aging, as well as acute inflammation, such as inflammatory bowel diseases of Crohn’s disease and ulcerative colitis. Ghrelin expression in both humans and mice under DIO is highly variable. In obese humans, ghrelin levels are decreased in plasma and serum, but increased in gastric tissues.50,5759 In obese mice, ghrelin levels are decreased in plasma, but increased in corpus and duodenum.5153 Interestingly, ghrelin expression in humans and mice is overall increased, which is evident in plasma and brain in aging, and colon and lymph nodes in Crohn’s disease and ulcerative colitis.43,48,60 GHS-R expression is decreased in hypothalamus and nodose ganglion of DIO mice. 49 GHS-R expression is mostly increased in pituitary, peritoneal fluid, colon, lymph node, and peripheral blood mononuclear cell (PBMC) in aging, Crohn’s disease, and ulcerative colitis.42,5961 The current knowledge of GHS-R expression in immune cells and its immune regulation is preliminary, which is the major reason that we write this review, hoping to inspire further investigation.

Table 2.

Expression of ghrelin and GHS-R in various human/mouse tissues under inflammatory states.

Expression Species Disease state Expression level change Time of sample collection or length of diet Tissue Detection method References
Ghrelin Human DIO 30 ± 7 years
30.4 ± 4.1 years
Plasma RIA, RT-PCR 50,57
DIO 44.3 ± 12.6 years Serum RT-PCR 58
DIO 44.3 ± 12.6 years Gastric RT-PCR 58
CD 30.5 ± 11 years Sigmoid colon RT-PCR 59
UC 39 ± 11.5 years Sigmoid colon RT-PCR 59
Mouse DIO HFD for 8–14 weeks Plasma RIA 51–53
DIO HFD for 7 weeks Corpus, duodenum RIA 51
Aging 4–22 months Plasma RIA 48
Aging 2–24 months Brain RT-PCR 43
Aging 24–28 months Brain RT-PCR 43
UC DSS for 7 days Lymph node RT-PCR 60
GHS-R Human CD 30.5 ± 11 years Sigmoid colon RT-PCR 59
CD 39 ± 11.5 years T cells from PBMC Flow cytometry 59
Mouse DIO HFD for 11 weeks Nodose ganglion, hypothalamus RT-PCR 49
Aging Highest in 1–2 months 1–2 months Pituitary RT-PCR 43
Aging 13–16 months Peritoneal fluid RT-PCR 42
UC DSS for 7 days Colon, lymph node RT-PCR 60,61

DIO: diet-induced obesity; CD: Crohn’s disease; UC: ulcerative colitis; HFD: high-fat diet; GHS-R: growth hormone secretagogue receptor; PBMC: peripheral blood mononuclear cell; DSS: dextran sulfate sodium; RT-PCR: reverse transcription polymerase chain reaction; RIA: radioimmunoassay.

The arrows note the expression level changes of ghrelin or GHS-R: ↑ indicates increase and ↓ indicates decrease.

GHS-R expression in innate and adaptive immune cells

In general, immune cells can be divided into two major categories: innate immune cells, which include monocytes, macrophages, neutrophils, DCs, and NK cells, and adaptive immune cells, which include T and B cells. 62 Here, we have summarized the current literature regarding the expression of GHS-R in innate and adaptive immune cells, its expression under inflammatory conditions, as well as reported functions of GHS-R in these immune cells.

Monocytes

GHS-R is reported to express in human monocytes. One study reported that GHS-R expression in monocytes is similar between Crohn’s disease patients and healthy controls. 59 Another study showed that 21% of CD14+ human monocytes in PBMC express GHS-R, and ghrelin administration reduces secretion of pro-inflammatory cytokines (interleukin [IL]-1b and IL-6) in lipopolysaccharide (LPS)-treated monocytes. 63

Macrophages

Ellie Metchnikoff identified macrophages as phagocytes that function to clear cellular debris and bacterial products; macrophages also repair tissue injury. Macrophages are essential in retaining tissue homeostasis; macrophage dysfunction triggers systemic inflammation. 64 Responding to microenvironmental cues, macrophages rapidly react to stimuli to combat insults/injury.65,66 Polarization is a hallmark phenotypical change of macrophages; the phenotypical change of macrophages directly affects their functional outcomes. Macrophages are often described as a dichotomy of M1-like and M2-like macrophages: M1-like macrophages being pro-inflammatory and M2-like macrophages being anti-inflammatory.6669 While the M1–M2 paradigm is widely used in vitro, 66 it is now recognized that in tissues, macrophages exist as a continuous spectrum instead of a distinctive M1–M2 population.70,71 For the convenience of the discussion, we will continue to use the M1–M2 concept in this review.

We previously reported that GHS-R is highly expressed in mouse peritoneal macrophages (PMs), 60% of the highest-expressing tissue, the hypothalamus. 42 Moreover, we found that GHS-R expression is elevated in PM of aged mice, and that ablation of GHS-R shifts macrophages of peritoneal fluid and white adipose tissue toward an anti-inflammatory state in the aged mice. 42 We also found that M1-like macrophages are decreased, and pro-inflammatory cytokine expression of tumor necrosis factor (TNF)-a, IL-1b, and IL-6 is reduced in epididymal fat of GHS-R-ablated mice fed high fructose corn syrup (HFCS). 55 Similar to our findings, others also showed that adipose tissue in GHS-R-ablated mice under DIO had decreased pro-inflammatory macrophage marker expressions including monocyte chemoattractant protein (MCP)-1, TNF-a, and inducible nitric oxide synthase (iNOS), whereas the anti-inflammatory markers of arginase-1 (Arg-1) and macrophage galactose-type lectin-1 (Mgl-1) were increased. 72 In addition, we reported that the macrophage Raw264.7 cell line expresses GHS-R, and GHS-R antagonist or GHS-R siRNA knockdown resulted in significant reduction of pro-inflammatory marker expressions under LPS-induced inflammation. 55 Collectively, these results indicate that GHS-R has a cell-autonomous effect in macrophages and GHS-R is involved in the polarization and immune remodeling of macrophages. That is to note that GHS-R, as G protein-coupled receptor, has high ligand-independent constitutive activity, which is about 50% of the maximal activity.73,74 The effect of GHS-R in immune cells could be mediated by both ligand-dependent and independent (constitutive) mechanisms. We observed that global deletion of GHS-R did not alter the serum ghrelin level compared to wild-type littermates under both fed and fast conditions. 18 While more studies are needed, we speculate that the effects of GHS-R in macrophages are mediated by both ghrelin-dependent and ghrelin-independent mechanisms. That is likely that even without endogenous ghrelin, GHS-R can modulate immune cells by altering its down-steam signaling pathways. Current studies reported by us and others on the effect of GHS-R on macrophage polarization are summarized in Table 3, showing that suppression of GHS-R promotes an anti-inflammatory polarization of macrophages both in vivo and in vitro.

Table 3.

GHS-R-associated changes of macrophage cell-surface markers and cytokine expression in mice.

Inflammatory stimuli Summary of results Tissues/cells Treatment or age Detection method References
HFD-induced obesity Adipose tissue macrophages of GHS-R global knockout mice show reduced M1 pro-inflammatory markers (MCP-1, TNF-a, and iNOS) and increased M2 anti-inflammatory markers (Arg-1 and Mgl-1). Epididymal adipose tissue 12-month feeding RT-PCR 72
HFCS-induced inflammation Ablation of GHS-R reduces pro-inflammatory cytokines of TNF-a, IL-1b, and IL-6, as well as chemokine MCP-1 in epididymal fat. Epididymal adipose tissue 10-month feeding RT-PCR, flow cytometry 55
Ablation of GHS-R reduces mRNA expression of F4/80, MCP-1, CD11c, TNF-a, and IL-1b in peritoneal macrophages. Peritoneal fluid 10-month feeding RT-PCR 55
Aging GHS-R ablation shifts the M1/M2 macrophage ratio toward anti-inflammatory and attenuates pro-inflammatory cytokines (F4/80, MCP-1, TNF-a, IL-1b, IL-6, CD11c) in epididymal white adipose tissue. Epididymal adipose tissue 13- to 16-month old RT-PCR 42
GHS-R ablation attenuates age-associated expression of F4/80, MCP-1, TNF-a, IL-1b, and CD11c in brown adipose tissue. Brown adipose tissue 13- to 16-month old RT-PCR 42
GHS-R ablation reduces the expression of MCP-1, TNF-a, IL-1b, and CD11c in the peritoneal macrophages, shifting peritoneal macrophages toward anti-inflammatory state. Peritoneal fluid 13- to 16-month old RT-PCR 42
LPS-induced inflammation GHS-R siRNA knockdown in RAW 264.7 cells results in decreased expression of MCP-1, CD11c, TNF-a, IL-1b, and IL-6. RAW264.7 18 h LPS (1 μg/mL) RT-PCR 55
GHS-R shRNA knockdown in RAW264.7 cells attenuates LPS-induced increases of MCP-1, TNF-a, IL-1b, and CD11c. RAW264.7 16 h LPS (1 μg/mL) RT-PCR 42
GHS-R antagonist attenuates LPS-induced increase of TNF-a and IL-1b. RAW264.7 16 h LPS (1 μg/mL) RT-PCR 42

GHS-R: growth hormone secretagogue receptor; HFCS: high fructose corn syrup; HFD: high-fat diet; LPS: lipopolysaccharide; MCP-1: monocyte chemoattractant protein; TNF: tumor necrosis factor; iNOS: inducible nitric oxide synthase; IL: interleukin; RT-PCR: reverse transcription polymerase chain reaction; RIA: radioimmunoassay.

DCs

While there are reports showing GHS-R expression in DCs,63,75,76 the function of GHS-R in DCs is still debatable. Dixit et al. reported that GHS-R is detected in both immature and mature monocyte-derived DCs. 63 GHS-R expression has been reported in thymic stromal cells, which include DCs, macrophages, and epithelial cells. 77 The same group also reported that the number of CD11c+ DCs in thymic medulla of aged mice is enhanced by ghrelin administration, and also showed co-localization of GHS-R and CD11c+ DCs, which suggest that age-associated decline of thymic output may be linked to reduced expression of thymic ghrelin and GHS-R. 75 However, it needs to note that the cell-specific function of GHS-R in DCs has not been determined.

Neutrophils

Human neutrophils from the blood have previously been found to express both ghrelin and GHS-R. 56 Currently, only limited information is available regarding whether GHS-R in neutrophils affects the function of neutrophils in humans and rodents. A study using vertebrate rainbow trout (Oncorhynchus mykiss) reported that ghrelin treatment in vitro elevates production of superoxide in phagocytic leukocytes from trout head kidney. 78 In the same study, GHS-R antagonist treatment suppressed ghrelin-induced superoxide production in phagocytic leukocytes; this suggests that GHS-R might play a role in oxidative burst of the phagosome. The precise function of GHS-R in neutrophils in humans and rodents remains to be further elucidated.

NK cells

Although very limited, human NK cells have been found to express both ghrelin and GHS-R. 79 In both lean and obese mice, ghrelin treatment has been shown to increase NK cells in peritoneal fluid of surgically induced sepsis. 80 Similarly, another study reported that ghrelin-treated rats with Trypanosoma cruzi infection had elevated NK cells in spleen. 81 The same study revealed that about 10% of NK cells in spleen express GHS-R in steady state, and the GHS-R expression is increased in activated NK cells. 81 However, it is not known if GHS-R alters function of NK cells.

B lymphocytes

B lymphocytes from human blood were found to express both ghrelin and GHS-R, whereas the expression level varies among individuals. 56 It is also shown that human leukemic B cell lines, including Raji and Daudi, express GHS-R. Currently, functional relevance of GHS-R in B lymphocytes is unclear.

T lymphocytes

Similar to B cells, human T lymphocytes from blood also express ghrelin and GHS-R and show great variability among individuals. 56 It was reported that the percentage of GHS-R positive CD3+ T cells from human blood is significantly elevated in Crohn’s disease patients compared to healthy controls, while the percentage of GHS-R expressing T cells in ulcerative colitis patients did not differ from controls. 59 Dixit et al. 82 showed that the active form of ghrelin is represented in more than 70% of human T cells. Furthermore, approximately 30% of highly purified human resting T cells express GHS-R, and its expression is upregulated under T cell activation. 63 In the same study, ghrelin treatment inhibits pro-inflammatory cytokine production (IL-1β and IL-6) in primary human T cells upon anti-CD3 activation. Interestingly, this study revealed that GHS-R is co-localized with aggregated lipid rafts under cellular activation, suggesting GHS-R may affect T cell activation.

In line with human data, murine splenic T cells are also reported to express GHS-R and ghrelin. Ghrelin administration restores proliferation of CD4+ T cells, showing a protective effect for surgically induced sepsis. 83 Similarly, it was reported that ghrelin treatment increases γδ T cells in peritoneal fluid of lean septic mice. 80 However, another study showed that ghrelin suppresses anti-CD3-induced proliferation of mouse splenic T cells in a dose-dependent manner. 84 Current literature in the area is confounding. One study showed that ghrelin does not affect proliferation of human T cells, nor their secretion of IL-2 and interferon gamma (IFN-γ). 63 However, another study reported that ghrelin treatment inhibits mRNA expression of Th1 cell-produced cytokines of IL-2 and IFN-γ, as well as Th2 cell-produced cytokines of IL-4 and IL-10 in the murine splenic T cells. 84 Thus, while there is evidence that ghrelin/GHS-R signaling has an important role in T cells, much more studies are needed to further define the precise function of GHS-R in T cells.

Conclusions

GHS-R is expressed in immune cells, and its expression changes dynamically depending on the microenvironment. GHS-R responds to various inflammatory cues, and is altered under various disease conditions such as DIO, aging, and inflammatory diseases.

While the role of GHS-R in the immune system is not fully understood, the current available data clearly indicate that GHS-R has a role in both innate and adaptive immunity. There is strong evidence that GHS-R is expressed in macrophages and T cells, and GHS-R is associated with macrophage functional programming and T cell activation. The loss-of-function study indicates that ablation of GHS-R shifts the macrophage phenotype from pro-inflammatory to anti-inflammatory, which supports a role of GHS-R in macrophage polarization. In addition, there is also evidence that GHS-R expression is upregulated in activated T cells and co-localized with lipid rafts. Increased expression levels of GHS-R in macrophages and T cells are detected in inflammatory disease models; this suggests that ghrelin/GHS-R may be involved in the pathogenesis of inflammatory diseases. Blockage of the GHS-R signaling pathway may have a therapeutic potential in both metabolic and inflammatory disease states. Besides macrophages and T cells, the other immune cells such as DCs, neutrophils, NK cells, and B cells also express GHS-R, whereas the function of GHS-R in these immune cells remains to be elucidated.

Overall, ghrelin signaling has exciting implications in immune functions in health and great therapeutic potentials in disease. However, it is important to note that the ghrelin signaling in immunity is likely to be complex. There are differential immune phenotypes exhibited by ghrelin and GHS-R, notably both ghrelin and GHS-R deficiency show anti-inflammatory effects. This discrepancy suggests that GHS-R may activate both ghrelin-dependent and ghrelin-independent signaling pathways in immune cells, which underscores the critical need for further investigation. Single-cell RNA sequencing and cell-specific gene targeting would be advantageous for a better understanding of the cell-specific roles and regulatory mechanisms of ghrelin signaling in immunity.

Footnotes

Authors’ Contributions: JYN and YS contributed to the conceptualization. JYN and MH contributed to the investigation. YS contributed to the resources. JYN and MH contributed to the writing—original draft preparation. JYN, XDT, BSP, GW, and YS contributed to the writing—review and editing. GW and YS contributed to the supervision. YS contributed to the project administration. YS and XDT contributed to the funding acquisition. All authors have read and agreed to the published version of the manuscript.

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by the National Institutes of Health (NIH) (R01DK118334 and R01AG064869) (Y.S.), as well as BrightFocus Foundation (A2019630S) (YS). This work was also supported, in part, by funding from the Texas A&M AgriLife Institute for Advancing Health Through Agriculture (YS), NIH (R01DK123826 and R01DK129960) (X-DT), and US Department of Veterans Affairs Merit Review Award (I01BX001690) (X-DT).

References

  • 1. Fang C, Xu H, Guo S, Mertens-Talcott SU, Sun Y. Ghrelin signaling in immunometabolism and inflamm-aging. Adv Exp Med Biol 2018;1090: 165–82 [DOI] [PubMed] [Google Scholar]
  • 2. Albarran-Zeckler RG, Sun Y, Smith RG. Physiological roles revealed by ghrelin and ghrelin receptor deficient mice. Peptides 2011;32:2229–35 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Lee JH, Lin L, Xu P, Saito K, Wei Q, Meadows AG, Bongmba OY, Pradhan G, Zheng H, Xu Y, Sun Y. Neuronal deletion of ghrelin receptor almost completely prevents diet-induced obesity. Diabetes 2016;65:2169–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Lin L, Saha PK, Ma X, Henshaw IO, Shao L, Chang BH, Buras ED, Tong Q, Chan L, McGuinness OP, Sun Y. Ablation of ghrelin receptor reduces adiposity and improves insulin sensitivity during aging by regulating fat metabolism in white and brown adipose tissues. Aging Cell 2011;10:996–1010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Pradhan G, Samson SL, Sun Y. Ghrelin: much more than a hunger hormone. Curr Opin Clin Nutr Metab Care 2013;16:619–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Wu C-S, Bongmba O, Yue J, Lee J, Lin L, Saito K, Pradhan G, Li D-P, Pan H-L, Xu A, Guo S, Xu Y, Sun Y. Suppression of GHS-R in AgRP neurons mitigates diet-induced obesity by activating thermogenesis. Int J Mol Sci 2017;18:832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Lin L, Lee J, Wang R, Wang R, Sheikh-Hamad D, Zang Q, Sun Y. aP2-cre mediated ablation of GHS-R attenuates adiposity and improves insulin sensitivity during aging. Int J Mol Sci 2018;19:3002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Ma X, Lin Y, Lin L, Qin G, Pereira FA, Haymond MW, Butte NF, Sun Y. Ablation of ghrelin receptor in leptin-deficient ob/ob mice has paradoxical effects on glucose homeostasis when compared with ablation of ghrelin in ob/ob mice. Am J Physiol Endocrinol Metab 2012;303:E422–131 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Sun Y. New insights on neuronal functions of ghrelin receptor GHS-R in obesity. J Neurol Neuromed 2018;3:69–74 [Google Scholar]
  • 10. Pradhan G, Lee JH, Wu C-S, Wang H, Lin L, Donti T, Graham BH, Rajan AS, Balasubramanyam A, Samson SL, Guo S, Sun Y. Mechanistic investigation of GHS-R mediated glucose-stimulated insulin secretion in pancreatic islets. Biomolecules 2022;12:407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Villarreal D, Pradhan G, Zhou Y, Xue B, Sun Y. Diverse and complementary effects of ghrelin and obestatin. Biomolecules 2022;12:517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Tokudome T, Otani K, Miyazato M, Kangawa K. Ghrelin and the heart. Peptides 2019;111:42–6 [DOI] [PubMed] [Google Scholar]
  • 13. Wu C-S, Wei Q, Wang H, Kim DM, Balderas M, Wu G, Lawler J, Safe S, Guo S, Devaraj S, Chen Z, Sun Y. Protective effects of ghrelin on fasting-induced muscle atrophy in aging mice. The Journals of Gerontology: Series A 2018;75:621–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. van der Velde M, van der Eerden BC, Sun Y, Almering JM, van der Lely AJ, Delhanty PJ, Smith RG, van Leeuwen JP. An age-dependent interaction with leptin unmasks Ghrelin’s bone-protective effects. Endocrinology 2012;153:3593–602 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Soleyman-Jahi S, Sadeghi F, Pastaki Khoshbin A, Khani L, Roosta V, Zendehdel K. Attribution of ghrelin to cancer; attempts to unravel an apparent controversy. Front Oncol 2019;9:1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Hosoda H, Kojima M, Mizushima T, Shimizu S, Kangawa K. Structural divergence of human ghrelin. Identification of multiple ghrelin-derived molecules produced by post-translational processing. J Biol Chem 2003;278:64–70 [DOI] [PubMed] [Google Scholar]
  • 17. Soares JB, Leite-Moreira AF. Ghrelin, des-acyl ghrelin and obestatin: three pieces of the same puzzle. Peptides 2008;29:1255–70 [DOI] [PubMed] [Google Scholar]
  • 18. Sun Y, Wang P, Zheng H, Smith RG. Ghrelin stimulation of growth hormone release and appetite is mediated through the growth hormone secretagogue receptor. Proc Natl Acad Sci U S A 2004;101:4679–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Kojima M, Kangawa K. Structure and function of ghrelin. Berlin; Heidelberg: Springer, pp. 89–115 [Google Scholar]
  • 20. Matsumoto M, Hosoda H, Kitajima Y, Morozumi N, Minamitake Y, Tanaka S, Matsuo H, Kojima M, Hayashi Y, Kangawa K. Structure-activity relationship of ghrelin: pharmacological study of ghrelin peptides. Biochem Biophys Res Commun 2001;287:142–6 [DOI] [PubMed] [Google Scholar]
  • 21. Bednarek MA, Feighner SD, Pong SS, McKee KK, Hreniuk DL, Silva MV, Warren VA, Howard AD, Van Der Ploeg LH, Heck JV. Structure-function studies on the new growth hormone-releasing peptide, ghrelin: minimal sequence of ghrelin necessary for activation of growth hormone secretagogue receptor 1a. J Med Chem 2000;43:4370–6 [DOI] [PubMed] [Google Scholar]
  • 22. Pradhan G, Wu CS, Han Lee J, Kanikarla P, Guo S, Yechoor VK, Samson SL, Sun Y. Obestatin stimulates glucose-induced insulin secretion through ghrelin receptor GHS-R. Sci Rep 2017;7:979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Granata R, Settanni F, Gallo D, Trovato L, Biancone L, Cantaluppi V, Nano R, Annunziata M, Campiglia P, Arnoletti E, Ghe C, Volante M, Papotti M, Muccioli G, Ghigo E. Obestatin promotes survival of pancreatic beta-cells and human islets and induces expression of genes involved in the regulation of beta-cell mass and function. Diabetes 2008;57:967–79 [DOI] [PubMed] [Google Scholar]
  • 24. Zhang JV, Ren PG, Avsian-Kretchmer O, Luo CW, Rauch R, Klein C, Hsueh AJ. Obestatin, a peptide encoded by the ghrelin gene, opposes ghrelin’s effects on food intake. Science 2005;310:996–9 [DOI] [PubMed] [Google Scholar]
  • 25. Lu X, Huang L, Huang Z, Feng D, Clark RJ, Chen C. LEAP-2: an emerging endogenous ghrelin receptor antagonist in the pathophysiology of obesity. Front Endocrinol (Lausanne) 2021;12:717544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Yin Y, Li Y, Zhang W. The growth hormone secretagogue receptor: its intracellular signaling and regulation. Int J Mol Sci 2014;15:4837–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Smith RG, Van der Ploeg LH, Howard AD, Feighner SD, Cheng K, Hickey GJ, Wyvratt MJ, Jr, Fisher MH, Nargund RP, Patchett AA. Peptidomimetic regulation of growth hormone secretion. Endocr Rev 1997;18:621–45 [DOI] [PubMed] [Google Scholar]
  • 28. Navarro G, Aguinaga D, Angelats E, Medrano M, Moreno E, Mallol J, Cortés A, Canela EI, Casadó V, McCormick PJ, Lluís C, Ferré S. A significant role of the truncated ghrelin receptor GHS-R1b in ghrelin-induced signaling in neurons. J Biol Chem 2016;291:13048–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Liu R, Nikolajczyk BS. Tissue immune cells fuel obesity-associated inflammation in adipose tissue and beyond. Front Immunol 2019;10:1587 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Mraz M, Haluzik M. The role of adipose tissue immune cells in obesity and low-grade inflammation. J Endocrinol 2014;222:R113–27 [DOI] [PubMed] [Google Scholar]
  • 31. Ponnappan S, Ponnappan U. Aging and immune function: molecular mechanisms to interventions. Antioxid Redox Sig 2011;14:1551–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Thomas R, Wang W, Su DM. Contributions of age-related thymic involution to immunosenescence and inflammaging. Immun Ageing 2020;17:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Jaiswal AK, Makhija S, Stahr N, Sandey M, Suryawanshi A, Saxena A, Dagur PK, McCoy JP, Levine SJ, Mishra A. Dendritic cell-restricted progenitors contribute to obesity-associated airway inflammation via Adam17-p38 MAPK-dependent pathway. Front Immunol 2020;11:363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Lu J, Zhao J, Meng H, Zhang X. Adipose tissue-resident immune cells in obesity and type 2 diabetes. Front Immunol 2019;10:1173 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Kiran S, Kumar V, Murphy EA, Enos RT, Singh UP. High fat diet-induced CD8(+) T cells in adipose tissue mediate macrophages to sustain low-grade chronic inflammation. Front Immunol 2021;12:680944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Pham TD, Chng MHY, Roskin KM, Jackson KJL, Nguyen KD, Glanville J, Lee JY, Engleman EG, Boyd SD. High-fat diet induces systemic B-cell repertoire changes associated with insulin resistance. Mucosal Immunol 2017;10:1468–79 [DOI] [PubMed] [Google Scholar]
  • 37. Bertola A, Ciucci T, Rousseau D, Bourlier V, Duffaut C, Bonnafous S, Blin-Wakkach C, Anty R, Iannelli A, Gugenheim J, Tran A, Bouloumie A, Gual P, Wakkach A. Identification of adipose tissue dendritic cells correlated with obesity-associated insulin-resistance and inducing Th17 responses in mice and patients. Diabetes 2012;61:2238–47 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Cho KW, Zamarron BF, Muir LA, Singer K, Porsche CE, DelProposto JB, Geletka L, Meyer KA, O’Rourke RW, Lumeng CN. Adipose tissue dendritic cells are independent contributors to obesity-induced inflammation and insulin resistance. J Immunol 2016;197:3650–61 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Bosco N, Noti M. The aging gut microbiome and its impact on host immunity. Genes Immun 2021;22:289–303 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 1999;402:656–60 [DOI] [PubMed] [Google Scholar]
  • 41. Gnanapavan S, Kola B, Bustin SA, Morris DG, McGee P, Fairclough P, Bhattacharya S, Carpenter R, Grossman AB, Korbonits M. The tissue distribution of the mRNA of ghrelin and subtypes of its receptor, GHS-R, in humans. J Clin Endocrinol Metab 2002;87:2988. [DOI] [PubMed] [Google Scholar]
  • 42. Lin L, Lee JH, Buras ED, Yu K, Wang R, Smith CW, Wu H, Sheikh-Hamad D, Sun Y. Ghrelin receptor regulates adipose tissue inflammation in aging. Aging (Albany NY) 2016;8:178–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Sun Y, Garcia JM, Smith RG. Ghrelin and growth hormone secretagogue receptor expression in mice during aging. Endocrinology 2007;148:1323–9 [DOI] [PubMed] [Google Scholar]
  • 44. Zigman JM, Jones JE, Lee CE, Saper CB, Elmquist JK. Expression of ghrelin receptor mRNA in the rat and the mouse brain. J Comp Neurol 2006;494:528–48 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Jeffery PL, Duncan RP, Yeh AH, Jaskolski RA, Hammond DS, Herington AC, Chopin LK. Expression of the ghrelin axis in the mouse: an exon 4-deleted mouse proghrelin variant encodes a novel C terminal peptide. Endocrinology 2005;146:432–40 [DOI] [PubMed] [Google Scholar]
  • 46. MacParland SA, Liu JC, Ma X-Z, Innes BT, Bartczak AM, Gage BK, Manuel J, Khuu N, Echeverri J, Linares I, Gupta R, Cheng ML, Liu LY, Camat D, Chung SW, Seliga RK, Shao Z, Lee E, Ogawa S, Ogawa M, Wilson MD, Fish JE, Selzner M, Ghanekar A, Grant D, Greig P, Sapisochin G, Selzner N, Winegarden N, Adeyi O, Keller G, Bader GD, McGilvray ID. Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations. Nature Communications 2018;9:4383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Freitas-Lopes MA, Mafra K, David BA, Carvalho-Gontijo R, Menezes GB. Differential location and distribution of hepatic immune cells. Cells 2017;6:48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Lin L, Lee JH, Bongmba OY, Ma X, Zhu X, Sheikh-Hamad D, Sun Y. The suppression of ghrelin signaling mitigates age-associated thermogenic impairment. Aging (Albany NY) 2014;6:1019–32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Naznin F, Toshinai K, Waise TM, NamKoong C, Md Moin AS, Sakoda H, Nakazato M. Diet-induced obesity causes peripheral and central ghrelin resistance by promoting inflammation. J Endocrinol 2015;226:81–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Tschop M, Weyer C, Tataranni PA, Devanarayan V, Ravussin E, Heiman ML. Circulating ghrelin levels are decreased in human obesity. Diabetes 2001;50:707–9 [DOI] [PubMed] [Google Scholar]
  • 51. Uchida A, Zechner JF, Mani BK, Park WM, Aguirre V, Zigman JM. Altered ghrelin secretion in mice in response to diet-induced obesity and Roux-en-Y gastric bypass. Mol Metab 2014;3:717–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Chandarana K, Gelegen C, Karra E, Choudhury AI, Drew ME, Fauveau V, Viollet B, Andreelli F, Withers DJ, Batterham RL. Diet and gastrointestinal bypass–induced weight loss. Diabetes 2011;60:810–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Moesgaard SG, Ahren B, Carr RD, Gram DX, Brand CL, Sundler F. Effects of high-fat feeding and fasting on ghrelin expression in the mouse stomach. Regul Pept 2004;120:261–7 [DOI] [PubMed] [Google Scholar]
  • 54. Shankar K, Takemi S, Gupta D, Varshney S, Mani BK, Osborne-Lawrence S, Metzger NP, Richard CP, Berglund ED, Zigman JM. Ghrelin cell-expressed insulin receptors mediate meal- and obesity-induced declines in plasma ghrelin. JCI Insight 2021;6:e146983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Ma X, Lin L, Yue J, Pradhan G, Qin G, Minze LJ, Wu H, Sheikh-Hamad D, Smith CW, Sun Y. Ghrelin receptor regulates HFCS-induced adipose inflammation and insulin resistance. Nutr Diabetes 2013;3:e99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Hattori N, Saito T, Yagyu T, Jiang BH, Kitagawa K, Inagaki C. GH, GH receptor, GH secretagogue receptor, and ghrelin expression in human T cells, B cells, and neutrophils. J Clin Endocrinol Metab 2001;86:4284–91 [DOI] [PubMed] [Google Scholar]
  • 57. Shiiya T, Nakazato M, Mizuta M, Date Y, Mondal MS, Tanaka M, Nozoe S, Hosoda H, Kangawa K, Matsukura S. Plasma ghrelin levels in lean and obese humans and the effect of glucose on ghrelin secretion. J Clin Endocrinol Metab 2002;87:240–4 [DOI] [PubMed] [Google Scholar]
  • 58. Ritze Y, Schollenberger A, Hamze Sinno M, Bühler N, Böhle M, Bárdos G, Sauer H, Mack I, Enck P, Zipfel S, Meile T, Königsrainer A, Kramer M, Bischoff SC. Gastric ghrelin, GOAT, leptin, and leptinR expression as well as peripheral serotonin are dysregulated in humans with obesity. Neurogastroenterol Motil 2016;28:806–15 [DOI] [PubMed] [Google Scholar]
  • 59. Hosomi S, Oshitani N, Kamata N, Sogawa M, Yamagami H, Watanabe K, Tominaga K, Watanabe T, Fujiwara Y, Maeda K, Hirakawa K, Arakawa T. Phenotypical and functional study of ghrelin and its receptor in the pathogenesis of Crohn’s disease. Inflamm Bowel Dis 2008;14:1205–13 [DOI] [PubMed] [Google Scholar]
  • 60. Liu ZZ, Wang WG, Li Q, Tang M, Li J, Wu WT, Wan YH, Wang ZG, Bao SS, Fei J. Growth hormone secretagogue receptor is important in the development of experimental colitis. Cell Biosci 2015;5:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Noh JY, Wu CS, DeLuca JAA, Devaraj S, Jayaraman A, Alaniz RC, Tan XD, Allred CD, Sun Y. Novel role of ghrelin receptor in gut dysbiosis and experimental colitis in aging. Int J Mol Sci 2022;23:2219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Hoebe K, Janssen E, Beutler B. The interface between innate and adaptive immunity. Nat Immunol 2004;5:971–4 [DOI] [PubMed] [Google Scholar]
  • 63. Dixit VD, Schaffer EM, Pyle RS, Collins GD, Sakthivel SK, Palaniappan R, Lillard JW, Jr, Taub DD. Ghrelin inhibits leptin- and activation-induced proinflammatory cytokine expression by human monocytes and T cells. J Clin Invest 2004;114:57–66 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Metchnikoff E. The prolongation of life; optimistic studies. London: Heinemann, 1907 [Google Scholar]
  • 65. Orliaguet L, Dalmas E, Drareni K, Venteclef N, Alzaid F. Mechanisms of macrophage polarization in insulin signaling and sensitivity. Front Endocrinol (Lausanne) 2020;11:62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Atri C, Guerfali F, Laouini D. Role of human macrophage polarization in inflammation during infectious diseases. Int J Mol Sci 2018;19:1801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Italiani P, Boraschi D. From monocytes to M1/M2 macrophages: phenotypical vs. functional differentiation. Front Immunol 2014;5:514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000prime Rep 2014;6:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Ley K. M1 means kill; M2 means heal. J Immunol 2017;199:2191–3 [DOI] [PubMed] [Google Scholar]
  • 70. Kang B, Alvarado LJ, Kim T, Lehmann ML, Cho H, He J, Li P, Kim BH, Larochelle A, Kelsall BL. Commensal microbiota drive the functional diversification of colon macrophages. Mucosal Immunol 2020;13:216–29 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol 2005;5:953–64 [DOI] [PubMed] [Google Scholar]
  • 72. Yuan F, Ma J, Xiang X, Lan H, Xu Y, Zhao J, Li Y, Zhang W. Improvement of adipose macrophage polarization in high fat diet-induced obese GHSR knockout mice. Biomed Res Int 2018;2018:4924325–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Holst B, Holliday ND, Bach A, Elling CE, Cox HM, Schwartz TW. Common structural basis for constitutive activity of the ghrelin receptor family. J Biol Chem 2004;279:53806–17 [DOI] [PubMed] [Google Scholar]
  • 74. Petersen PS, Woldbye DP, Madsen AN, Egerod KL, Jin C, Lang M, Rasmussen M, Beck-Sickinger AG, Holst B. In vivo characterization of high Basal signaling from the ghrelin receptor. Endocrinology 2009;150:4920–30 [DOI] [PubMed] [Google Scholar]
  • 75. Dixit VD, Yang H, Sun Y, Weeraratna AT, Youm YH, Smith RG, Taub DD. Ghrelin promotes thymopoiesis during aging. J Clin Invest 2007;117:2778–90 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Dixit VD, Taub DD. Ghrelin and immunity: a young player in an old field. Exp Gerontol 2005;40:900–10 [DOI] [PubMed] [Google Scholar]
  • 77. Youm Y-H, Yang H, Sun Y, Smith RG, Manley NR, Vandanmagsar B, Dixit VD. Deficient ghrelin receptor-mediated signaling compromises thymic stromal cell microenvironment by accelerating thymic adiposity. J Biol Chem 2009;284:7068–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Yada T, Kaiya H, Mutoh K, Azuma T, Hyodo S, Kangawa K. Ghrelin stimulates phagocytosis and superoxide production in fish leukocytes. J Endocrinol 2006;189:57–65 [DOI] [PubMed] [Google Scholar]
  • 79. Uhlen M, Fagerberg L, Hallstrom BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson A, Kampf C, Sjostedt E, Asplund A, Olsson I, Edlund K, Lundberg E, Navani S, Szigyarto CA, Odeberg J, Djureinovic D, Takanen JO, Hober S, Alm T, Edqvist PH, Berling H, Tegel H, Mulder J, Rockberg J, Nilsson P, Schwenk JM, Hamsten M, von Feilitzen K, Forsberg M, Persson L, Johansson F, Zwahlen M, von Heijne G, Nielsen J, Ponten F. Proteomics. Tissue-based map of the human proteome. Science 2015;347:1260419. [DOI] [PubMed] [Google Scholar]
  • 80. Siegl D, Midura EF, Annecke T, Conzen P, Caldwell CC, Tschoep J. The effect of ghrelin upon the early immune response in lean and obese mice during sepsis. PLoS ONE 2015;10:e0122211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. de Paula Silva F, da Costa CMB, Pereira LM, Lessa DFS, Pitol DL, Issa JPM, do Prado, Junior JC, Abrahao AAC. Effects of ghrelin supplementation on the acute phase of Chagas disease in rats. Parasit Vectors 2019;12:532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Dixit VD, Yang H, Cooper-Jenkins A, Giri BB, Patel K, Taub DD. Reduction of T cell-derived ghrelin enhances proinflammatory cytokine expression: implications for age-associated increases in inflammation. Blood 2009;113:5202–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Zhou M, Aziz M, Ochani M, Yang WL, Sharma A, Wang P. The protective role of human ghrelin in sepsis: restoration of CD4 T cell proliferation. PLoS ONE 2018;13:e0201139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Xia Q, Pang W, Pan H, Zheng Y, Kang J-S, Zhu S-G. Effects of ghrelin on the proliferation and secretion of splenic T lymphocytes in mice. Regulatory Peptides 2004;122:173–8 [DOI] [PubMed] [Google Scholar]

Articles from Experimental Biology and Medicine are provided here courtesy of Frontiers Media SA

RESOURCES