Hostname: page-component-6bb9c88b65-x9fsb Total loading time: 0 Render date: 2025-07-21T18:41:40.102Z Has data issue: false hasContentIssue false

Amino acid metabolism-mediated immune cell fate and function in pigs

Published online by Cambridge University Press:  11 April 2025

Liuqin He
Affiliation:
Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan international joint laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, China Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
Shunshun Jin
Affiliation:
Department of Animal Science, University of Manitoba, Winnipeg, Manitoba, Canada
Tiejun Li
Affiliation:
Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
Yulong Yin*
Affiliation:
Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
*
Corresponding author: Yulong Yin; Email: yinyulong@isa.ac.cn
Rights & Permissions [Opens in a new window]

Abstract

Amino acids are fundamental to sustaining life. They are crucial for intracellular processes, such as energy metabolism, biosynthesis of nucleotides, and maintenance of oxidative homeostasis. These processes ensure the proper functioning of cells (including immune cells) and organs. Many studies have demonstrated that immune cells, as key players in immune regulation, have distinct amino acid demands, and their rapid growth and activation are shaped by amino acid availability in their microenvironment. In particular, the proliferation, maturation, and functional responses of innate immune cells are closely linked to amino acid metabolism. The transport, sensing, and mobilization of amino acids drive metabolic reprogramming to support these processes. Therefore, this review focuses on the influence of amino acids on the fate and function of immune cells across development, homeostasis, activation, and effector phases, highlighting the underlying mechanisms. It provides a scientific basis for improving disease resistance and production efficiency in animals.

Information

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2025. Published by Cambridge University Press on behalf of Zhejiang University and Zhejiang University Press.

Introduction

Amino acids are essential components that not only condense into peptides and proteins but also maintain the homeostasis of the immune system. Their influence on immune cell fate and functionality is multifaceted (Tomé Reference Tomé2021). On the one hand, amino acids provide the essential structural components and energy sources required for immune cell proliferation, differentiation, and functioning. On the other hand, immune cells have specific demands for amino acids. Processes like amino acid mobilization, uptake, and sensing drive metabolic reprogramming in immune cells, which affects their fate and functionality (Hope and Salmond Reference Hope and Salmond2021; Pearce and Pearce Reference Pearce and Pearce2013). Studies have revealed that metabolic pathways of various immune cell types during origin, proliferation, differentiation, maturation, activation, and senescence differ significantly from those in resting states. For example, during an immune response to infections or environmental changes, immune cells transition into a highly active state characterized by elevated expression of amino acid transporters (Song et al. Reference Song, Li and Tao2020). T cells serve as a representative example. When they become activated, they rapidly proliferate and upregulate the transcription and translation of key immune-related genes. This heightened activity accelerates amino acid metabolism to support the synthesis of essential macromolecules like proteins and nucleotides (Kaech et al. Reference Kaech, Hemby and Kersh2002).

Emerging research has revealed the link between dysregulated amino acid metabolism and various pathological conditions, such as metabolic disorders and immune dysfunction. In animal husbandry, amino acids are important feed components for livestock and poultry, effectively regulating immune dysfunction triggered by external or internal factors, thereby influencing disease resistance and survival rates (Bai and Plastow Reference Bai and Plastow2022). This is especially relevant under policies restricting antibiotic use and in the context of African swine fever, where amino acids, as key nutritional regulators, have become increasingly important in improving health and immune function in livestock such as pigs. Previous studies have suggested that serine and glutamine can influence the porcine T cells activity, improving host defense against pathogens (Chen et al. Reference Chen, Xia and He2020; Ma et al. Reference Ma, Zhang and Li2022; Ren et al. Reference Ren, Rajendran and Zhao2018; Shan et al. Reference Shan, Hu and Ni2022; Yu et al. Reference Yu, Tu and Yin2022; Zheng et al. Reference Zheng, Zhu and Zhao2023a). Research on amino acid metabolism pathways further highlights their role in regulating redox balance, gene expression in immune cells, and lymphocyte proliferation (Han et al. Reference Han, Ge and Ho2021; He et al. Reference He, Liu and Guan2023; Muri and Kopf Reference Muri and Kopf2021; Zhang et al. Reference Zhang, Wang and Yin2020). For example, arginine supports the growth and proliferation of immune cells while contributing to the synthesis of key immune mediators, such as nitric oxide and cytokines, which are vital for modulating inflammation and controlling autoimmune disorders (Martíi Líndez and Reith Reference Martíi Líndez and Reith2021). These findings illustrate the dynamic interplay between amino acid metabolism and immune cells, driven by signal transduction and metabolic reprogramming, which adapts to physiological and pathological conditions in animals. This provides an essential theoretical foundation for an in-depth understanding of how amino acid metabolism affects the immune cell fate and function and also opens new ways for the development of nutritional strategies to enhance animal immunity. Therefore, this review aims to focus on three key areas: (1) the mechanisms by which immune cells sense and selectively utilize amino acids; (2) the influence of amino acids on metabolic reprogramming in immune cells; and (3) the impact of amino acids on the function and fate of immune cells. Finally, we discuss future directions for research on how amino acid metabolism impacts immune cells, aiming to establish a theoretical basis that fosters integration between immunology and nutrition and expands the existing nutritional theories on amino acids.

Sensing and uptake of amino acids by immune cells

Mechanisms of amino acid sensing in immune cells

Rapamycin, a compound isolated from soil, exhibits antifungal and antitumor properties (Ghoname et al. Reference Ghoname, Ghozlan and Sabry2022). In mammals, the mammalian target of rapamycin (mTOR) is a conserved serine/threonine kinase that senses environmental changes to regulate eukaryotic cell metabolism and growth (Mota-Martorell et al. Reference Mota-Martorell, Jove and Pradas2020). This kinase forms two distinct complexes: mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). The primary difference between these complexes lies in their unique scaffold proteins, with mTORC1 showing higher sensitivity to macrolide drugs like rapamycin (Fu et al. Reference Fu, Fu and Su2023). Activation of mTORC1 involves key metabolites, such as amino acids, glucose, and nucleotides, through signaling pathways involving small GTPases, such as Ras homolog enriched in brain (RHEB) GTPases (Kim and Kim Reference Kim and Kim2016; Nguyen et al. Reference Nguyen, Frank and Jewell2017; Zhu and Wang Reference Zhu and Wang2020). At the molecular level, the Rag complex, consisting of heterodimers formed by RagA or RagB with RagC or RagD, collaborates with the Ragulator complex, which acts as a guanine nucleotide exchange factor for RagC or RagD. Together, these complexes facilitate the lysosomal translocation of mTORC1, a critical step that enables its activation by RHEB on the lysosomal surface (Groenewoud Marlous and Zwartkruis Fried Reference Groenewoud Marlous and Zwartkruis Fried2013; Lama-Sherpa et al. Reference Lama-Sherpa, Jeong and Jewell2023; Sancak et al. Reference Sancak, Bar-Peled and Zoncu2010; Tsujimoto et al. Reference Tsujimoto, Takamatsu and Kumanogoh2023). The regulatory mechanism of the Rag complex is further refined by the GTPase activating protein activity toward Rags 1 (GATOR1), which specifically targets RagA and RagB, modulating their activity (Shen et al. Reference Shen, Valenstein and Gu2019). Its localization to the lysosomal membrane is mediated through its interaction with the Rag complex. This process is further regulated by the KICSTOR complex, a key assembly that includes SZT2, and by the GTPase activating protein activity toward Rags 2 (GATOR2) complex, which acts upstream to fine-tune the Rag complex activity (Cui et al. Reference Cui, Joiner and Jansen2023; Valenstein et al. Reference Valenstein, Lalgudi and Gu2024; Zhao et al. Reference Zhao, Guan and Xu2023).

Some studies have demonstrated that immune cells sense amino acids through mTORC1, which relies on vacuolar-type ATPase on lysosomes (Wang et al. Reference Wang, Schianchi and Neumann2021d). Under conditions of sufficient amino acid availability, the vacuolar-type ATPase activates the guanine nucleotide exchange function of Ragulator, facilitating nucleotide exchange and activating Rag GTPases (Brady et al. Reference Brady, Diab and Puertollano2016; Hertel et al. Reference Hertel, Alves and Dutz2022). The activated Rag GTPases then recruit mTORC1 to the lysosomal membrane, positioning it near RHEB, which triggers mTORC1 activation (Carroll Reference Carroll2020; Gan et al. Reference Gan, Seki and Shen2019; Sancak et al. Reference Sancak, Bar-Peled and Zoncu2010, Reference Sancak, Peterson and Shaul2008). Following its activation, mTORC1 localizes to the lysosome, where it phosphorylates 4EBP1. This phosphorylation event releases eIF4E, which initiates protein synthesis (Battaglioni et al. Reference Battaglioni, Benjamin and Wälchli2022; Clemens et al. Reference Clemens, Elia and Morley2013; Grosso et al. Reference Grosso, Pesce and Brina2011; Wang et al. Reference Wang, Lei and Zhang2022). GATOR2 can inhibit GATOR1, but when arginine or leucine is present, the cellular arginine sensor for mTORC1 (CASTOR1) or Sestrin can bind GATOR2 in response to arginine or leucine, which relieves this inhibition (Jiang et al. Reference Jiang, Dai and He2023). Additionally, some studies have shown that a sensor of S-adenosylmethionine (SAM) (a metabolic product of methionine) upstream of mTORC1 (SAMTOR) can suppress mTORC1 by interacting with GATOR1, and binding of SAM to SAMTOR destroys this interaction (Kitada et al. Reference Kitada, Xu and Ogura2020). Amino acids such as arginine and methionine play important roles in activating mTORC1 across various T-cell subsets (Abdullah et al., Reference Abdullah, Zeng and Margerum2022). Key transporters, including L-type amino acid transporter 1 (LAT-1/SLC7A5) and SLC1A5, are critical for initiating mTORC1 signaling in naive, activated, and regulatory T cells (Tregs) (Grosso et al. Reference Grosso, Pesce and Brina2011; Huang et al. Reference Huang, Long and Zhou2020; Yang et al. Reference Yang, Lu and Piao2022). During the first division of CD8+ T cells, the asymmetric distribution of amino acid transporters changes how much mTOR accumulates in proximal and distal daughter cells, which eventually determines whether CD8+ T cells become memory cells or effector cells (Arsenio et al. Reference Arsenio, Kakaradov and Metz2014, Reference Arsenio, Metz and Chang2015; Cai et al. Reference Cai, Li and Wang2022; Chen et al. Reference Chen, Xu and Sun2023; Pollizzi et al. Reference Pollizzi, Sun and Patel2016). Additionally, the Rag complex is indispensable for detecting amino acid levels and has been found to suppress regulatory T-cell function (Delmonte et al. Reference Delmonte, Villa and Notarangelo2020; Shi et al. Reference Shi, Chapman and Wen2019; Thangavelu et al. Reference Thangavelu, Andrejeva and Bolivar-Wagers2022; Zhu et al. Reference Zhu, Wu and Li2024). Thus, immune cells sense environmental amino acid levels by regulating mTORC1 activity, reshaping their fate and functionality.

Immune cells also sense amino acids through general control nonderepressible 2 (GCN2), which detects tRNA that is not fully loaded with amino acids (Kim et al. Reference Kim, Sundrud and Zhou2020). Under normal conditions, tRNA that carries amino acids accumulates at ribosomes during protein translation, ensuring that the growing peptide chain receives enough amino acids (Englander et al. Reference Englander, Avins and Fleisher2015). When amino acids are scarce, uncharged or unloaded tRNA accumulates in the cell. This accumulation leads to an overall slowdown in protein translation to save energy and resources, accompanied by a selective reduction in the translation of mRNAs that restore cellular homeostasis (Darnell et al. Reference Darnell, Subramaniam and O’Shea2018). Excessive accumulation of uncharged tRNAs interacts with GCN2, causing a structural rearrangement that triggers downstream signaling pathways (Wu et al. Reference Wu, Zinshteyn and Wehner2019). Upon activation, GCN2 phosphorylates eIF2α at serine 51, which disrupts the assembly of the eIF2/tRNAiMet/GTP ternary complex, a crucial step in initiating protein translation (Kedersha et al. Reference Kedersha, Chen and Gilks2002) (Fig. 1). Previous studies have suggested that GCN2 activation negatively affects T-cell proliferation and Treg differentiation (Rashidi et al. Reference Rashidi, Miska and Lee-Chang2020; Sonner et al. Reference Sonner, Deumelandt and Ott2016; Zheng et al. Reference Zheng, Yao and Ge2023b). Studies on amino acid deprivation, such as those using indoleamine 2,3-dioxygenase to degrade tryptophan, have revealed that indoleamine 2,3-dioxygenase increases IL-10 production while reducing IL-12 expression in macrophages via a GCN2-dependent mechanism that inhibits protein synthesis (Battu et al. Reference Battu, Minhas and Mishra2017; Yan et al. Reference Yan, Zhang and Gran2010). During apoptosis or exposure to apoptotic antigens, macrophages suppress IL-12 mRNA expression while increasing IL-10 transcript translation (Filardy et al. Reference Filardy, Pires and Nunes2010). Additionally, the amino acid starvation response activated by halofuginone can suppress IL-1β production in macrophages through GCN2 activation (Battu et al. Reference Battu, Afroz and Giddaluru2018). Collectively, immune cells can sense amino acids through both the GCN2 and mTORC1 pathways, which allows them to regulate their fate and function based on amino acid availability.

Figure 1. Mechanism of amino acid sensing by immune cells. When amino acid levels are low, uncharged tRNA activates GCN2, which interferes with the recruitment of mTORC1 substrates and blocks protein synthesis. In contrast, when amino acid levels are sufficient, cytoplasmic amino acid sensors inhibit GATOR1 through GATOR2, ultimately activating mTORC1 and promoting protein synthesis.

Mechanisms of amino acid mobilization and uptake in immune cells

Amino acid mobilization and uptake are critical regulatory points that impact the significantly influence immune cell function and fate. These cells primarily acquire amino acids through transporters that move amino acids from the microenvironment into the cells (Almeida et al. Reference Almeida, Lochner and Berod2016; Ma et al. Reference Ma, Ming and Wu2024; Tae et al. Reference Tae, Kim and S.-w.2023; Yang et al. Reference Yang, Chu and Liu2023). Activation of the T-cell receptor (TCR) has been shown to trigger metabolic reprogramming in T cells, leading to changes in glycolysis, oxidative phosphorylation, and fatty acid β-oxidation pathways (Cammann et al. Reference Cammann, Rath and Reichl2016; Marelli-Berg et al. Reference Marelli-Berg, Fu and Mauro2012; Patsoukis et al. Reference Patsoukis, Bardhan and Weaver2016; Xuekai et al. Reference Xuekai, Yan and Jian2024). These metabolic changes rely heavily on amino acid availability and the corresponding transporters. The large amino acid transporter 1, also referred to as SLC7A5, forms a heterodimeric complex with the transmembrane protein CD98 (also known as SLC3A2) to transport large hydrophobic amino acids (Bröer et al. Reference Bröer, Gauthier-Coles and Rahimi2019). This transporter facilitates the uptake of seven essential amino acids, excluding lysine and threonine (Brunocilla et al. Reference Brunocilla, Console and Rovella2023). T-cell activation dramatically increases the expression of the LAT-1/CD98 complex, supporting antigen recognition and rapid clonal expansion (Cantor and Ginsberg Reference Cantor and Ginsberg2012; Hayashi et al. Reference Hayashi, Jutabha and Endou2013). Some studies have found that CD4+ T cells with a knockout of the Slc7a5 gene exhibit impaired antigen responses, with an inability to proliferate or differentiate into Th1 and Th17 subsets (Sinclair et al. Reference Sinclair, Rolf and Emslie2013; Yeramian et al. Reference Yeramian, Martin and Serrat2006). These findings suggest the critical role of LAT-1 not only in early T-cell activation but also in guiding T-cell differentiation. In animal models of lupus and psoriasis induced by imiquimod (a TLR7 agonist), the absence or inhibition of LAT-1 significantly reduces IL-17 secretion and suppress the expansion of γδ T cells and CD4⁺ T cells (Zhang et al. Reference Zhang, Zhang and Yu2021b). Other amino acid transporters, such as SLC7A7, which forms complexes with CD98, are responsible for the transport of lysine, arginine, and other amino acids, highlighting the diversity and complexity of amino acid transporters in immune function regulation (Dai et al. Reference Dai, Feng and Hu2021). Once amino acids enter immune cells, the cells can recycle certain chemical groups to synthesize new amino acids (Yang et al. Reference Yang, Chu and Liu2023). Lysosomes within immune cells also contain amino acid transport mechanisms as part of the self-protection system during starvation (Wang et al. Reference Wang, Wan and Du2021a). These lysosomal transporters are also crucial for immune functions such as the production of type I interferons in dendritic cells and immunoglobulin G synthesis in B cells (Akkaya et al. Reference Akkaya, Akkaya and Miozzo2017; Jiang et al. Reference Jiang, Yan and Wang2018; Li et al. Reference Li, Wu and Jiang2022; Montoya et al. Reference Montoya, Schiavoni and Mattei2002).

The asymmetric distribution of amino acid transporters is crucial in determining T-cell fate (Arsenio et al. Reference Arsenio, Kakaradov and Metz2014). When interacting with antigen-presenting cells, SLC7A5 displays a notable asymmetric distribution (Kedl et al. Reference Kedl, Rees and Hildeman2000). This distribution results in differing amino acid concentrations and metabolic activities between proximal and distal T-cell subsets, ultimately influencing immune cell function and fate (Pollizzi et al. Reference Pollizzi, Sun and Patel2016). In proximal daughter cells, high SLC7A5 expression promotes increased amino acid uptake, particularly glutamine, which supports rapid proliferation and high energy demand (Huang et al. Reference Huang, Wang and Hong2023). Increased glutamine enhances glycolysis, providing abundant energy and carbon sources for biosynthesis. Additionally, glutamine contributes to α-ketoglutarate production, accelerating the tricarboxylic acid (TCA) cycle and enhancing metabolic activity and energy generation (Oh et al. Reference Oh, Sun and Zhao2020). Enhanced glycolysis and metabolic activity activate the c-Myc and mTORC1 signaling pathways, laying the molecular foundation for proximal daughter cells to differentiate into effector T cells. In contrast, distal daughter cells with lower SLC7A5 expression exhibit reduced amino acid uptake and metabolic activity, resulting in weaker metabolic activity. While this situation does not favor rapid cell proliferation, it does help the cells conserve energy and resources, preparing them for future reactivation and rapid responses. Consequently, these distal daughter cells tend to develop into memory T cells that survive long-term and can respond quickly when they encounter the same antigen again (Kaech and Cui Reference Kaech and Cui2012; Montacchiesi and Pace Reference Montacchiesi and Pace2022; Pollizzi et al. Reference Pollizzi, Sun and Patel2016; Verbist et al. Reference Verbist, Guy and Milasta2016). Overall, the uptake of amino acids and the asymmetric distribution of their transporters are decisive factors in T-cell function and fate, affecting both immediate immune responses and long-term functionality and survival.

T-cell development

The development of T cells occurs primarily in the thymus. Hematopoietic stem cells derived from the bone marrow differentiate into common lymphoid progenitor cells, which migrate to the thymus and further develop into progenitor T cells (Karsunky et al. Reference Karsunky, Inlay and Serwold2008). These progenitor cells undergo TCR rearrangement and selection processes, giving rise to mature conventional αβ T cells as well as unconventional subsets such as γδ T cells, natural killer T cells (NKT), mucosal-associated invariant T cells, and thymic-derived regulatory T cells (tTreg) (Hoebeke et al. Reference Hoebeke, De Smedt and Stolz2007; Liu et al. Reference Liu, Wang and Ding2015). T-cell maturation progresses through distinct developmental stages, including double-negative (DN) and double-positive (DP) phases. The DN stage is further classified into DN1, DN2a, DN2b, DN3a, DN3b, and DN4 phases (Guha et al. Reference Guha, Bhuniya and Shukla2020; Wang et al. Reference Wang, Qi and Yao2021b; Yui et al. Reference Yui, Feng and Rothenberg2010). The transition from DN2 to DN3 determines whether pro-T cells differentiate into αβ or γδ T cells (Ciofani and Zúñiga-Pflücker Reference Ciofani and Zúñiga-Pflücker2010). Pre-TCR and Notch signaling in DN3a cells are crucial for β-selection and the subsequent development of conventional αβ T cells (Ciofani and Zúñiga-Pflücker Reference Ciofani and Zúñiga-Pflücker2010). Conversely, DN2 and DN3 cells exposed to elevated interleukin-7 (IL-7) signaling pathways are driven towards the γδ T-cell lineage (Saba et al. Reference Saba, Kosan and Vassen2011). DP cells have the potential to differentiate into unconventional subsets, such as iNKT and tTreg cells (Wang et al. Reference Wang, Zhao and Jin2021c; Winter and Krueger Reference Winter and Krueger2019) (Fig. 2). In pigs, the composition of T-cell subsets differs significantly from other mammals. Some studies have shown that during late gestation in sows, the population of γδ T cells in embryonic blood and peripheral lymphoid tissues increases sharply, far exceeding the number of CD4⁺CD8⁻ and CD4⁻CD8⁺ T lymphocytes. This αβ-to-γδ T-cell ratio is distinct from what is observed in other mammals (Augustyniak et al. Reference Augustyniak, Czyżewska-Dors and Pomorska-Mól2023; Bianchi et al. Reference Bianchi, Zwart and Jeurissen1992; Le Page et al. Reference Le Page, Baldwin and Telfer2022; Schalk et al. Reference Schalk, Pfaffinger and Schmucker2019). Some T cells in pigs may originate outside the thymus (Licence and Binns Reference Licence and Binns1995). Previous studies identified mitotic T cells in the gastrointestinal epithelium of pigs, referred to as intraepithelial T cells (IEK) (Wiarda et al. Reference Wiarda, Trachsel and Bond2020). Currently, research on unconventional T cells in pigs remains limited. Further studies on these cells could enhance our understanding of their roles in amino acid metabolism, disease prevention, and vaccine responses.

Figure 2. Development of T cells. T-cell development proceeds through a series of stages. DN1 cells can differentiate into B cells, myeloid cells, and innate T cells, while DN2b and DN3a cells can give rise to γδ T cells. At the DN3 stage, the pre-TCR complex-formed by TCRβ, pTα, and CD3 molecules-promotes β selection and drives the transition from DN3 to DN4. Both the pre-TCR and Notch signals are crucial for β selection and for the shift from the DN to the DP stage. After positive selection in the thymic cortex and negative selection in the thymic medulla, DP cells eventually differentiate into CD4+ T cells, CD8+ T cells, or iNKT cells.

Immune cell metabolism

Once immune cells mature, they leave the thymus. When T cells are in a resting state and have not been activated by receptor engagement or cytokine signals, their metabolic demands remain low. They rely mainly on fatty acid β-oxidation and TCA cycle. The maintenance of this resting metabolic state in T cells requires external signals such as IL-7. When T cells encounter an antigen, they undergo dynamic metabolic, differentiating into effector T cells in a process known as metabolic reprogramming. The activated T cells switch their primary energy source from oxidative phosphorylation to glycolysis (Almeida et al. Reference Almeida, Lochner and Berod2016; Kempkes et al. Reference Kempkes, Joosten and Koenen2019). Although glycolysis produces less ATP per cycle compared to oxidative phosphorylation, it can generate ATP at a faster rate, work effectively in low-oxygen or acidic environments, and provide higher biosynthetic efficiency to help maintain redox balance (Choudhury Reference Choudhury2021; Lees et al. Reference Lees, Gardner and Harvey2017; Wang et al. Reference Wang, Guan and Wang2020). Metabolites produced during glycolysis feed into the pentose phosphate pathway, which contributes to amino acid and nucleotide biosynthesis and generates nicotinamide adenine dinucleotide phosphate for reducing power (Ge et al. Reference Ge, Yang and Zhou2020). During the later stages of immune responses, a small subset of antigen-specific T cells persists as long-lived memory T cells (Xu et al. Reference Xu, Bhanumathy and Wu2016). These memory T cells are characterized by an increased mitochondrial mass, which enhances their spare respiratory capacity and prepares them for rapid responses upon re-exposure to antigens (Li and Zhang Reference Li and Zhang2020). Therefore, amino acid metabolism is an integral component of T-cell metabolism, providing intermediates that support these metabolic pathways and determining immune cell functionality (Almeida et al. Reference Almeida, Lochner and Berod2016; Kelly and Pearce Reference Kelly and Pearce2020; Wang and Zou Reference Wang and Zou2020) (Fig. 3).

Figure 3. Amino acid metabolism in activated T cell. Serine is required for the production of cytokines in activated T cells with the help of the key glycolytic enzyme PKM2. Although pyruvate can be used to make alanine, activated T cells reduce the synthesis of alanine from pyruvate in order to conserve pyruvate metabolism and convert it into acetyl-CoA for TCA cycle activity. Branched-chain amino acids (BCAAs) provide the TCA cycle with the intermediate product CoA. Glutamine and leucine also contribute to the TCA cycle via glutamate to α-ketoglutarate.

In addition to the metabolic distinctions between effector and memory T cells, various T-cell subsets exhibit unique metabolic characteristics. The differentiation of Th1, Th2, Th17, and Tregs is heavily influenced by cytokines such as interferon-γ (IFN-γ), interleukin-4, interleukin-6, and transforming growth factor-β, respectively (Barnes and Powrie Reference Barnes and Powrie2009; Liu et al. Reference Liu, Cao and Feng2015; Li et al. Reference Li, Spolski and Liao2014, Reference Li, Wan and Flavell2007a; Maloy et al. Reference Maloy, Salaun and Cahill2003; Marie et al. Reference Marie, Letterio and Gavin2005; O’Shea et al. Reference O’Shea, Steward-Tharp and Laurence2009; Tang et al. Reference Tang, Wang and Xing2020). While Th1, Th2, and Th17 cells primarily rely on glycolysis for energy and biosynthesis, Tregs employ a mixed metabolic approach that integrates glycolysis, fatty acid oxidation, and oxidative phosphorylation (Kempkes et al. Reference Kempkes, Joosten and Koenen2019; Ma et al. Reference Ma, Ming and Wu2024) Interestingly, shifting the metabolic balance can alter T-cell differentiation. Inhibiting glycolysis during Th17 differentiation has been shown to favor the generation of Tregs (Barbi et al. Reference Barbi, Pardoll and Pan2013; Shi et al. Reference Shi, Wang and Huang2011; Sun et al. Reference Sun, Fu and Zhou2017; Zhang et al. Reference Zhang, Gang and Yang2021a). Similarly, the addition of exogenous fatty acids to T-cell cultures significantly suppresses the production of cytokines associated with Th1, Th2, and Th17 cells, while having minimal impact on Tregs. Notably, this suppression of effector T cells by fatty acids cannot be reversed by the addition of cytokines that typically promote their differentiation (Allen et al. Reference Allen, Fan and Monk2014; Michalek et al. Reference Michalek, Gerriets and Jacobs2011). Moreover, suppressing mTOR signaling to enhance fatty acid oxidation increases the number of memory T cells (Chi Reference Chi2012; Pearce et al. Reference Pearce, Walsh and Cejas2009; Waickman and Powell Reference Waickman and Powell2012). These findings underscore the fundamental metabolic differences between effector T cells and Tregs. Therefore, the study on amino acid metabolism-mediated T-cell fate and function has emerged as a hot and active topic in the present and future.

The impact of amino acid metabolism on immune cell fate and function

Glutamine metabolism-mediated immune cell fate

Glutamine and its metabolism are critical for the proliferation, differentiation, and activation of T cells by providing essential energy (Carr et al. Reference Carr, Kelman and Wu2010; Yaqoob and Calder Reference Yaqoob and Calder1997). Some studies have shown that in glutamine-free media, T cells predominantly differentiate into Tregs rather than Th17 cells, and prolonged glutamine deprivation exacerbates this effect (Edwards et al. Reference Edwards, Ngwa and Raybuck2021). Immune T cells rely on SLC38A2 to transport extracellular glutamine into the cell. Without this transporter, Th17 differentiation is significantly reduced, whereas the formation of Tregs remains unaffected (Zheng et al. Reference Zheng, Yao and Ge2023b). Within immune cells, glutamine metabolism requires the catalytic activity of glutaminase-1 (Gls1) to convert glutamine into glutamate. Th17 cells exhibit higher Gls1 expression levels than other T-cell subsets, with elevated intracellular levels of glutamate and α-ketoglutarate (a glutamate metabolite) (Kono et al. Reference Kono, Yoshida and Maeda2018). Previous studies have suggested that Gls1 deficiency reduces α-ketoglutarate levels, affecting chromatin states and gene expression, further inhibiting mTORC1 and IL-2 signaling, thereby impairing Th17 differentiation while promoting Th1 cell formation (Johnson et al. Reference Johnson, Wolf and Madden2018; Lin et al. Reference Lin, Ren and Xiong2024). The dependence of Th17 cells on glutamine has been further demonstrated in Gls1-deficient mice. Inhibition of Gls1, either through pharmacological means or siRNA-mediated approaches, significantly reduces Th17 differentiation in vitro (Kono et al. Reference Kono, Yoshida and Maeda2018).

Glutamine also influences the activation, proliferation, and development of other immune cells. In M1 macrophages, it enhances the production of α-ketoglutarate, a key metabolite that drives the synthesis of IL-1β and supports inflammatory responses (Kolliniati et al. Reference Kolliniati, Ieronymaki and Vergadi2021; Palmieri et al. Reference Palmieri, Gonzalez-Cotto and Baseler2020). In M2 macrophages, glutamine contributes to the TCA cycle, supporting anti-inflammatory responses (Gupta and Sarangi Reference Gupta and Sarangi2023; Jha et al. Reference Jha, Huang and Sergushichev2015; Liu et al. Reference Liu, Wang and Li2017; Zhao et al. Reference Zhao, Wang and Liu2020b). Additionally, glutamine is vital for the activation and functionality of NKT cells and B cells. NKT cells use glutamine to synthesize glutathione and hexosamines, while B cells depend on it for antibody production (Jiang et al. Reference Jiang, Yan and Wang2018; Loftus et al. Reference Loftus, Assmann and Kedia-Mehta2018). Overall, glutamine serves as a versatile metabolite that sustains the functionality and metabolic adaptability of diverse immune cell populations.

Branched-chain amino acids metabolism-mediated immune cell fate

In addition to glutamine, branched-chain amino acids (BCAAs) contribute to the TCA cycle and are involved in various biosynthetic processes. Through the generation of intermediates such as acetyl-CoA and succinyl-CoA, BCAAs provide essential substrates for the TCA cycle (Bo and Fujii Reference Bo and Fujii2025; Dimou et al. Reference Dimou, Tsimihodimos and Bairaktari2022; Neinast et al. Reference Neinast, Jang and Hui2019). During immune cell activation, neutral amino acid transporters, particularly SLC7A5/CD98, are significantly upregulated, serving as crucial mediators during pathogen infections and immune responses. In T cells, infections trigger elevated expression of SLC7A5, a process further maintained by IL-2 to ensure a constant supply of BCAAs (Almutairi et al. Reference Almutairi, Ali and He2019; Kang et al. Reference Kang, Song and Lee2024; Sinclair et al. Reference Sinclair, Rolf and Emslie2013). The inhibition of SLC7A5 has been shown to significantly reduce IFN-γ and IL-17 production, impairing the differentiation of Th1 and Th17 cells, while having minimal impact on the Tregs (Hayashi et al. Reference Hayashi, Jutabha and Endou2013; Song et al. Reference Song, Li and Tao2020). Loss of SLC7A5 in T cells also impairs mTORC1 and Myc-dependent glycolysis, affecting T-cell activation and proliferation (Marchingo et al. Reference Marchingo, Sinclair and Howden2020). In macrophages, lipopolysaccharide (LPS)-induced inflammatory responses also activate BCAA transporters, with leucine transported by SLC7A5 being essential for glycolysis (Yoon et al. Reference Yoon, Oh and Kang2018). Similarly, CD98 expression is closely linked to the proliferation of immune cells and cytokine production. Plasma cells with high CD98 expression demonstrate stronger immune responses, longer lifespans, and increased antibody production (Cantor et al. Reference Cantor, Browne and Ruppert2009, Reference Cantor, Slepak and Ege2011; Jensen et al. Reference Jensen, Potempa and Gotthardt2017; Nguyen et al. Reference Nguyen, Duan and Ali2021; Robinson et al. Reference Robinson, Dowling and Pitt2022). The enzyme BCAA aminotransferase 1 (BCAT1) is integral to BCAA metabolism, catalyzing the conversion of BCAAs into branched-chain keto acids, which serve as precursors for TCA cycle entry. Inhibition of BCAT1 effectively reduces glycolysis and oxidative phosphorylation, thereby reducing the production of anti-inflammatory metabolites (Papathanassiu et al. Reference Papathanassiu, Ko and Imprialou2017). These metabolic pathways not only drive short-term immune responses but also underpin the adaptive immune responses required for prolonged antigen exposure.

Serine metabolism-mediated immune cell fate

Serine regulates immune cell metabolic reprogramming, influencing their fate and function. It interacts with pyruvate kinase M2 (PKM2), enhancing glycolytic flux to provide energy for immune cells (Chaneton et al. Reference Chaneton, Hillmann and Zheng2012). Previous study has suggested that PKM2 activation in lipopolysaccharide-induced macrophages drives a shift toward glycolysis and speeds up IL-1β production (Bahiraii et al. Reference Bahiraii, Brenner and Yan2022; Palsson-mcdermott et al. Reference Palsson-mcdermott, Curtis Anne and Goel2015; Xie et al. Reference Xie, Yu and Kang2016). In CD4⁺ T cells, activation of the TCR promotes the nuclear translocation of PKM2, increasing glycolysis and facilitating differentiation into Th1 and Th17 subsets. Inhibiting PKM2 nuclear translocation can limit the differentiation of these cells and reduce cytokine production, which helps slow the progression of multiple sclerosis (Angiari et al. Reference Angiari, Runtsch and Sutton2020; Puckett et al. Reference Puckett, Alquraishi and Chowanadisai2021; Traba et al. Reference Traba, Sack and Waldmann2021). Additionally, limiting serine intake can reduce PKM2 activity, thereby lowering macrophage activation in atherosclerosis and decreasing LPS-induced IL-1β production (Liu et al. Reference Liu, Le and Chen2021; Shirai et al. Reference Shirai, Nazarewicz and Wallis2016; Tabas and Bornfeldt Reference Tabas and Bornfeldt2020). Similar findings from our lab have demonstrated that LPS-treated piglet serum exhibits elevated IL-1β levels, which are reduced by dietary serine supplementation (Zhou et al. Reference Zhou, Zhang and He2017). In a dextran sulfate sodium-induced colitis model, our study also indicated that adding serine lowered the levels or activity of proinflammatory cytokines in mice (Zhang et al. Reference Zhang, Hua and Zhang2018). Additional research highlights the role of phosphoglycerate dehydrogenase (PHGDH), a rate-limiting enzyme in the serine de novo synthesis pathway, in macrophages. Inhibiting PHGDH increases NAD⁺ levels and enhances the activity of NAD⁺-dependent SIRT1/3, which promotes IL-1β production. PHGDH also supports Toll-like receptor 4 transcription via H3K9/27 acetylation and activates the NLRP3 inflammasome by facilitating acetylation of inflammasome components (Wang et al. Reference Wang, Chen and Chen2024). Lack of serine can suppress IL-1β production in macrophages by inhibiting mTOR signaling (Chen et al. Reference Chen, Xia and He2020; Shan et al. Reference Shan, Hu and Ni2022). Moreover, our team was the first to discover that adding appropriate amounts of serine to sow diets during late pregnancy and lactation significantly increased antibody levels in both sows and piglets, raised the positive rate of CD4⁺/CD8⁺ cells, improved the growth performance of nursing piglets and the reproductive performance of sows, and enhanced immunity in both sows and piglets (He et al. Reference He, Liu and Long2020). This suggests that serine may be a promising new feed additive for improving swine immunity. Additionally, serine supports mitochondrial metabolism through serine hydroxy methyltransferase 2 (Shmt2), essential for mitochondrial translation and respiration. Shmt2-deficient models exhibit severe respiratory defects (Tani et al. Reference Tani, Ohnishi and Shitara2018). Serine-derived one-carbon units are crucial for nucleotide synthesis and methionine cycling, particularly in proliferating tissues. In Shmt2 null Jurkat cells, supplying one-carbon units can fix defects in mitochondrial respiration and translation, especially under low-glucose conditions, highlighting the vital role of this pathway in adjusting metabolic states (Minton et al. Reference Minton, Nam and McLaughlin2018). Moreover, serine is indispensable for T-cell proliferation and differentiation. The serine de novo synthesis pathway supports purine synthesis and one-carbon metabolism, while activated T cells upregulate key enzymes in this pathway to regulate immune responses and metabolism directly (Ma et al. Reference Ma, Bantug and Griss2017). In models of Pasteurella multocida infection, serine levels in the lungs significantly decrease. Supplementing serine in these mice reduces bacterial colonization and inflammatory responses, further demonstrating its critical role in immune regulation (He et al. Reference He, Yin and Wu2019).

Sulfur-containing amino acids metabolism-mediated immune cell fate

Methionine plays multiple roles in regulating immune cell function and fate, primarily through its involvement in methylation processes. By providing SAM, methionine drives the methylation of biomolecules, which can promote or inhibit transcription by changing how DNA is accessed by transcriptional machinery. During T-cell activation, both repressive and activating histone methylation events occur frequently, facilitating transcriptional remodeling (Henning et al. Reference Henning, Roychoudhuri and Restifo2018; Sinclair et al. Reference Sinclair, Howden and Brenes2019). Additionally, RNA methylation, particularly N6-methyladenosine modification, plays a pivotal role in maintaining T-cell homeostasis. The absence of this modification leads to defects in mRNA stability, splicing, and translation initiation, ultimately impairing T-cell proliferation and differentiation (Frye et al. Reference Frye, Harada and Behm2018; Galloway et al. Reference Galloway, Kaskar and Ditsova2021; Li et al. Reference Li, Tong and Zhu2017). Beyond transcriptional regulation, methionine metabolism has a profound impact on immune memory formation and function. The methylation of histones, RNA, and other cellular components relies on the availability of SAM. For example, Th17 cells starved of methionine or subjected to methionine cycle inhibition exhibit reduced H3K4 methylation, leading to decreased IL-17 production, while methionine restriction in Th1 cells similarly reduces IFN-γ expression. Additionally, dietary methionine restriction has been shown to reduce the number of IL-17- and IFN-γ-producing cells, thereby influencing immune responses in conditions such as experimental autoimmune encephalomyelitis (Roy et al. Reference Roy, Chen and Mamane2020).

Methionine not only participates directly in methylation reactions through its metabolite SAM but also supports immune cell function and proliferation through byproducts such as S-adenosylhomocysteine and further metabolic pathways. β-glucan-trained human peripheral blood mononuclear cells exhibit increased H3K4 trimethylation at cytokine and immune signaling gene promoters, enhancing cytokine production upon Candida albicans re-exposure (Quintin et al. Reference Quintin, Saeed and Martens Joost2012). Similarly, memory CD4⁺ T cells show enriched histone marks associated with cytokines such as IL-17 and IFN-γ and transcription factors like T-bet, establishing a “primed” chromatin state that enables rapid cytokine production upon stimulation (Durek et al. Reference Durek, Nordström and Gasparoni2016; Schmidl et al. Reference Schmidl, Delacher and Huehn2018). Methionine transport is critical for these processes. Following antigen stimulation, T cells upregulate methionine transporters, including SLC7A5, to meet the increased demand for methyl donor production and protein synthesis, which are essential for T-cell differentiation and function. The balance between SAM and its byproduct, S-adenosylhomocysteine, also modulates histone methylation levels. Accumulation of S-adenosylhomocysteine due to disruptions in methionine metabolism can inhibit histone methylation, further suppressing immune gene expression. This metabolic-epigenetic interplay ensures that immune cells efficiently integrate nutrient availability with functional adaptation, reinforcing the role of methionine metabolism in maintaining long-term immune memory (Kelly and Pearce Reference Kelly and Pearce2020).

Cysteine, another sulfur-containing amino acid, is vital for maintaining redox balance and methylation regulation. It serves as a key precursor for glutathione (GSH), one of the body’s main antioxidants that control reactive oxygen species levels and preserve intracellular redox balance. In immune cells, methionine metabolism can generate cysteine. Beyond the regulatory roles methionine plays, cysteine also modulates immune cell responses through specific mechanisms: (1) antioxidant and cellular protection: Upon activation, T cells, B cells, and macrophages upregulate GSH synthesis, with cysteine providing the sulfur atom essential for this process. For example, LPS-stimulated macrophages produce high ROS levels, which are neutralized by GSH to prevent oxidative damage. (2) sulfur metabolism and translational support: Cysteine also contributes to iron-sulfur cluster synthesis, vital for mitochondrial electron transport chains and various metabolic enzymes. These clusters play a vital role in sustaining energy production and metabolic activity in T cells and macrophages (Fig. 4). Additionally, cysteine plays a role in post-translational modifications. The sulfur from cysteine is critical for tRNA thiolation, which facilitates efficient translation, particularly for proteins required in immune activation. Furthermore, iron-sulfur clusters derived from cysteine support mitochondrial metabolism and electron transport chain function, ensuring energy production for highly active immune cells. Therefore, sulfur-containing amino acids play indispensable roles in regulating oxidative stress, cellular metabolism, epigenetic control, and long-term immune memory.

Figure 4. Sulfur-containing amino acids maintain redox status homeostasis in T cells. In T cells, methionine can be converted into cysteine. During this process, the intermediate S-adenosylmethionine donates methyl groups that modify immune effector proteins and nucleic acids, promoting cytokine gene expression in T cells and supporting innate and adaptive immune memory. Serine can also be transformed into cysteine, which contributes to the synthesis of the antioxidant glutathione and the formation of iron-sulfur (FeS) clusters.

Arginine metabolism-mediated immune cell fate

Arginine influences protein structure modifications and immune cell activity regulation through its metabolism. In the presence of SAM, arginine generates polyamines, highlighting the interdependence and centrality of arginine and SAM in immune cell metabolism (Puleston et al. Reference Puleston, Buck and Klein Geltink2019). Polyamine production signals sufficient nutrient supply, coordinates biosynthesis, and supports immune cell proliferation. Polyamines participate in producing rare amino acid derivatives critical for the post-translational modification of eukaryotic translation initiation factor 5a (eIF5a), a key regulator of translation elongation and termination (Pelechano and Alepuz Reference Pelechano and Alepuz2017; Schuller et al. Reference Schuller, Wu and Dever2017). Inhibition of the polyamine-eIF5A-taillessin pathway suppresses oxidative phosphorylation-dependent M2 macrophage polarization, leading to a shift towards glycolysis-dependent M1 polarization (Puleston et al. Reference Puleston, Buck and Klein Geltink2019).

Other amino acids metabolism-mediated on immune cell fate

During immune cell proliferation and differentiation, nucleotides synthesis relies on amino acids such as aspartate and glycine. Aspartate and serine-derived glycine provide carbon backbones for nucleotide formation (Lane and Fan Reference Lane and Fan2015). Glycine plays a particularly critical role in the early stages of T-cell activation (Ma et al. Reference Ma, Bantug and Griss2017). Although glycine can convert to alanine, T cells typically use alanine for protein synthesis rather than glycolysis (Ron-Harel et al. Reference Ron-Harel, Ghergurovich and Notarangelo2019). Tryptophan’s metabolic product, melatonin, can influence macrophage M1/M2 polarization (Xia et al. Reference Xia, Chen and Zeng2019). Additionally, melatonin regulates the activation and differentiation of T cells (Th17, Treg, and memory T cells) by activating calcineurin and the ERK1/2-C/EBPα signaling pathways (Ren et al. Reference Ren, Liu and Chen2017a). Gamma-aminobutyric acid promotes the differentiation of intestinal Th17 cells and the expression of IL-17 during E. coli infection (Ren et al. Reference Ren, Yin and Xiao2017b). These functions, along with the broader immunomodulatory roles of amino acids, have been extensively summarized in previous reviews (Bhandage and Barragan Reference Bhandage and Barragan2021; Castellano and Molinier-Frenkel Reference Castellano and Molinier-Frenkel2020; Liao et al. Reference Liao, Fan and Bin2022; Stone and Williams Reference Stone and Williams2023; Tantawy and Naguib Reference Tantawy and Naguib2019; Yang et al. Reference Yang, Chu and Liu2023; Zhao et al. Reference Zhao, Raines and Huang2020a).

Challenges and practical considerations in pig farming

Despite the potential benefits of amino acid supplementation in improving immune function and growth performance in pigs, several challenges must be addressed for successful application in pig farming. Amino acid bioavailability is a key determinant of immune cell function and fate, as immune cells require optimal amino acid availability to support proliferation, cytokine production, and immune signaling pathways (Yang and Liao Reference Yang and Liao2019). The absorption efficiency of amino acids depends on feed formulation, processing methods, and the presence of anti-nutritional factors. Arginine and glutamine play essential roles in immune regulation, supporting T-cell activation and macrophage function, but their bioavailability is highly dependent on feed processing and dietary composition (Fanimo et al. Reference Fanimo, Susenbeth and Südekum2006; Sá et al. Reference Sá, Moreno and Carciofi2020; Wu et al. Reference Wu, Bazer and Johnson2018). Feed formulation influences amino acid digestibility through ingredient selection, balancing essential and non-essential amino acids to meet metabolic demands (Buraczewska et al. Reference Buraczewska, Wasilewko and Fandrejewski1999). Processing methods, such as extrusion and pelleting, can enhance amino acid bioavailability by breaking down cell walls and denaturing anti-nutritional proteins, but excessive heat treatment may degrade heat-sensitive amino acids like lysine and methionine (Ohh et al. Reference Ohh, Han and Chae2002). Anti-nutritional factors, such as tannins and phytates in plant-based feedstuffs, can inhibit amino acid absorption by forming complexes with proteins, reducing enzymatic hydrolysis and intestinal uptake (Woyengo and Nyachoti Reference Woyengo and Nyachoti2013). Plant-derived protein sources contain fiber and phytates, which can lower amino acid digestibility and reduce their availability to immune cells, thereby impairing immune function (Myrie et al. Reference Myrie, Bertolo and Sauer2008). Pigs (13-35 kg) fed a low-protein diet (13.9% crude protein) also impair T-cell activation and proliferation, suggesting that protein restriction can compromise immune resilience (Peng et al. Reference Peng, Hu and Liu2016).

Amino acid metabolism also interacts with other nutrients, influencing immune homeostasis and inflammatory responses. Amino acid antagonism is a significant factor influencing nutrient bioavailability and immune regulation. For instance, excessive lysine intake can interfere with arginine absorption by competing for the same transporters, potentially reducing nitric oxide production and impairing macrophage-mediated immune responses (Li et al. Reference Li, Yin and Li2007b). Similarly, an imbalance in BCAAs, particularly excessive leucine, can suppress isoleucine and valine uptake. Valine deficiency can reduce lymphocyte proliferation and hinder the growth of lymphoid tissue (Wang et al. Reference Wang, Peng and Zhang2023). Deficiencies in key vitamins, such as vitamin B6, which is involved in amino acid metabolism, can impair amino acid transamination processes, leading to reduced protein synthesis and weakened immune responses (Li et al. Reference Li, Yin and Wang2019).

Although amino acid fortification enhances immune resilience and growth performance in pigs, its cost-effectiveness remains a significant consideration. While synthetic amino acid supplements can improve the bioavailability of nutrients, they can increase feed costs, so the cost-effectiveness must be carefully evaluated (Ju et al. Reference Ju, Yun and Jang2008). Future research should focus on optimizing dietary formulations that balance immune support with economic feasibility to maximize health and productivity outcomes in pig farming. Addressing these challenges through optimized feed strategies and targeted research will improve the practical implementation of amino acid-based nutritional interventions in pig farming, ensuring that immune cell function and fate are optimally regulated to improve disease resistance and overall health outcomes.

Gut microbiota, amino acid metabolism, and immune cell function in pigs

Gut microbes not only utilize dietary amino acids for microbial protein synthesis but also influence host amino acid availability through protein degradation and nitrogen recycling (Dai et al. Reference Dai, Wu and Zhu2011; Macfarlane and Macfarlane Reference Macfarlane and Macfarlane2012). Specific commensal bacteria, such as Bacteroides, Clostridium, Lactobacillus, and Streptococcus, contribute to protein fermentation and enhance amino acid bioavailability by synthesizing essential amino acids that the host cannot produce (Dalmasso et al. Reference Dalmasso, Nguyen and Yan2008). In the small intestine, microbial protein synthesis is predominant, whereas in the large intestine, amino acid catabolism dominates, generating various metabolites such as ammonia, short-chain fatty acids and biogenic amines. These metabolites not only impact on intestinal health but also modulate host immune responses (Blachier et al. Reference Blachier, Mariotti and Huneau2007; Collins et al. Reference Collins, Surette and Bercik2012).

Tryptophan is metabolized by gut bacteria into indole derivatives, such as indole-3-aldehyde, which activate the aryl hydrocarbon receptor in immune cells. Aryl hydrocarbon receptor activation induces IL-22 production, which enhances mucosal immunity and maintains gut homeostasis by promoting epithelial barrier integrity (Zelante et al. Reference Zelante, Iannitti and Cunha2013). Additionally, Tryptophan metabolism leads to the formation of kynurenine, a metabolite that modulates immune responses by interacting with dendritic cells and macrophages, affecting cytokine secretion and T-cell differentiation (Ma et al. Reference Ma, Guo and Zhang2018). Glutamine is a critical amino acid for immune cells and intestinal barrier function. Gut bacteria metabolize glutamine into glutamate, which influences the gut-microbiome-immune axis by supporting intestinal epithelial renewal and regulating immune responses (Blachier et al. Reference Blachier, Boutry and Bos2009). Arginine metabolism by gut microbiota also plays a key role in immune modulation. Certain bacteria convert arginine into ornithine and nitric oxide, both of which contribute to macrophage activation and pathogen clearance. Nitric oxide, produced by inducible nitric oxide synthase, is essential for immune defense, as it enhances macrophage antimicrobial activity and controls inflammatory responses (Kan et al. Reference Kan, Lee and Wilson2015). However, when gut microbiota composition is imbalanced, amino acid metabolism can generate harmful metabolites that impair immune function. For example, the overgrowth of proteolytic bacteria, such as certain Clostridium species, can lead to excessive production of ammonia and hydrogen sulfide, which negatively affect intestinal epithelial integrity and immune homeostasis (Blachier et al. Reference Blachier, Mariotti and Huneau2007). However, there is currently little understanding of the basic mechanisms of the interactions between amino acids, the microbiome and immune cells. Future research should clarify their relationship in order to improve pig health and productivity.

Conclusion and perspectives

In recent decades, the relationship between amino acid metabolism and T-cell development and function has gained increasing attention. Although significant progress has been made, many fundamental issues remain unresolved. A deeper understanding is needed to elucidate how amino acid metabolism shapes the fate and function of immune cells across different mammalian species, particularly in livestock and poultry, where research remains relatively limited. While numerous studies have explored the impact of amino acids on T-cell activity and differentiation, the role of amino acid metabolism in maintaining or reprogramming effector cells is still not fully understood. Some studies have confirmed that dietary amino acids must be digested in the intestine and then undergo intracellular transformations before the body can utilize them. Simply adding amino acids to the diet may not allow them to be directed to specific sites to exert their corresponding effects. Immune cells exhibit distinct responses to amino acid perturbations, emphasizing the need to decode metabolic requirements in various tissues and physiological contexts. Another major challenge is converting our knowledge of how amino acids affect immune cell fate and function into precise and targeted therapies. Future studies targeting amino acid-specific signaling pathways in immune cells under different conditions will facilitate the development of targeted metabolic strategies for combating pathogens, tumors, and related diseases.

Acknowledgements

This work was supported by the Key Fundamental Research Program of Hunan Province (2024JC0007), the National Natural Science Foundation of China (U23A20233, 32172755), the Hunan Science and Technology Innovation leading Talent Support Program (2023RC1054), Municipal scientific and technological innovation cooperation project of Changchun (24SH16), the Shandong Province Taishan Industry Leading Talents Project Blue Talents Project, and the China Agriculture Research System of MOF and MARA (CARS-35).

Author contributions

L.H. and S.J. contributed equally to this work. L.H. and S.J. wrote the manuscript. T.L. and Y.Y. edited the paper. All authors provided feedback on the writing.

Conflict of interests

The authors declare no competing interests.

Footnotes

#

These authors contributed equally to this article.

References

Abdullah, MO, Zeng, RX, Margerum, CL et al. (2022) Mitochondrial hyperfusion via metabolic sensing of regulatory amino acids. Cell Reports 40(7), 111198.Google Scholar
Akkaya, M, Akkaya, B, Miozzo, P et al. (2017) B cells produce type 1 IFNs in response to the TLR9 agonist CpG-A conjugated to cationic lipids. Journal of Immunology 199(3), 931940.Google Scholar
Allen, MJ, Fan, -Y-Y, Monk, JM et al. (2014) n–3 PUFAs reduce T-Helper 17 cell differentiation by decreasing responsiveness to interleukin-6 in isolated mouse splenic CD4+ T cells. The Journal of Nutrition 144(8), 13061313.Google Scholar
Almeida, L, Lochner, M, Berod, L et al. (2016) Metabolic pathways in T cell activation and lineage differentiation. Seminars in Immunology 28(5), 514524.Google Scholar
Almutairi, SM, Ali, AK, He, W et al. (2019) Interleukin-18 up-regulates amino acid transporters and facilitates amino acid–induced mTORC1 activation in natural killer cells. Journal of Biological Chemistry 294(12), 46444655.Google Scholar
Angiari, S, Runtsch, MC, Sutton, CE et al. (2020) Pharmacological activation of pyruvate kinase M2 inhibits CD4+ T cell pathogenicity and suppresses autoimmunity. Cell Metabolism 31(2), 391405.Google Scholar
Arsenio, J, Kakaradov, B, Metz, PJ et al. (2014) Early specification of CD8+ T lymphocyte fates during adaptive immunity revealed by single-cell gene-expression analyses. Nature Immunology 15(4), 365372.Google Scholar
Arsenio, J, Metz, PJ and Chang, JT (2015) Asymmetric cell division in T lymphocyte fate diversification. Trends in Immunology 36(11), 670683.Google Scholar
Augustyniak, A, Czyżewska-Dors, E and Pomorska-Mól, M (2023) Immune status of piglets during the first week of life: current knowledge, significance and assessment – a review. Annals of Animal Science 23(2), 391403.Google Scholar
Bahiraii, S, Brenner, M, Yan, F et al. (2022) Sulforaphane diminishes moonlighting of pyruvate kinase M2 and interleukin 1β expression in M1 (LPS) macrophages. Frontiers in Immunology 13, 935692.Google Scholar
Bai, X and Plastow, GS (2022) Breeding for disease resilience: opportunities to manage polymicrobial challenge and improve commercial performance in the pig industry. CABI Agriculture and Bioscience 3(1), 6.Google Scholar
Barbi, J, Pardoll, D and Pan, F (2013) Metabolic control of the Treg/Th17 axis. Immunological Reviews 252(1), 5277.Google Scholar
Barnes, MJ and Powrie, F (2009) Regulatory T cells reinforce intestinal homeostasis. Immunity 31(3), 401411.Google Scholar
Battaglioni, S, Benjamin, D, Wälchli, M et al. (2022) mTOR substrate phosphorylation in growth control. Cell 185(11), 18141836.Google Scholar
Battu, S, Afroz, S, Giddaluru, J et al. (2018) Amino acid starvation sensing dampens IL-1β production by activating riboclustering and autophagy. PLoS Biology 16(4), e2005317.Google Scholar
Battu, S, Minhas, G, Mishra, A et al. (2017) Amino acid sensing via general control nonderepressible-2 kinase and immunological programming. Frontiers in Immunology 8, 311557.Google Scholar
Bhandage, AK and Barragan, A (2021) GABAergic signaling by cells of the immune system: more the rule than the exception. Cellular and Molecular Life Sciences 78(15), 56675679.Google Scholar
Bianchi, ATJ, Zwart, RJ, Jeurissen, SHM et al. (1992) Development of the B- and T-cell compartments in porcine lymphoid organs from birth to adult life: an immunohistological approach. Veterinary Immunology and Immunopathology 33(3), 201221.Google Scholar
Blachier, F, Boutry, C, Bos, C et al. (2009) Metabolism and functions of l-glutamate in the epithelial cells of the small and large intestines. The American Journal of Clinical Nutrition 90(3), 814S821S.Google Scholar
Blachier, F, Mariotti, F, Huneau, JF et al. (2007) Effects of amino acid-derived luminal metabolites on the colonic epithelium and physiopathological consequences. Amino Acids 33(4), 547562.Google Scholar
Bo, T and Fujii, J (2025) Primary roles of branched chain amino acids (BCAAs) and their metabolism in physiology and metabolic disorders. Molecules 30(1), 56.Google Scholar
Brady, OA, Diab, HI and Puertollano, R (2016) Rags to riches: amino acid sensing by the Rag GTPases in health and disease. Small GTPases 7(4), 197206.Google Scholar
Bröer, A, Gauthier-Coles, G, Rahimi, F et al. (2019) Ablation of the ASCT2 (SLC1A5) gene encoding a neutral amino acid transporter reveals transporter plasticity and redundancy in cancer cells. Journal of Biological Chemistry, 294(11), 40124026.Google Scholar
Brunocilla, C, Console, L, Rovella, F et al. (2023) Insights into the transport cycle of LAT1 and interaction with the inhibitor JPH203. International Journal of Molecular Sciences, 24(4), 4042.Google Scholar
Buraczewska, L, Wasilewko, J, Fandrejewski, H et al. (1999) Formulation of pig diets according to ileal digestible amino acid content. Livestock Production Science, 59(1), 1324.Google Scholar
Cai, X, Li, H, Wang, M et al. (2022) mTOR participates in the formation, maintenance, and function of memory CD8+T cells regulated by glycometabolism. Biochemical Pharmacology 204, 115197.Google Scholar
Cammann, C, Rath, A, Reichl, U et al. (2016) Early changes in the metabolic profile of activated CD8+ T cells. BMC Cell Biology 17(1), 28.Google Scholar
Cantor, J, Browne, CD, Ruppert, R et al. (2009) CD98hc facilitates B cell proliferation and adaptive humoral immunity. Nature Immunology 10(4), 412419.Google Scholar
Cantor, JM and Ginsberg, MH (2012) CD98 at the crossroads of adaptive immunity and cancer. Journal of Cell Science 125(Pt 6), 13731382.Google Scholar
Cantor, J, Slepak, M, Ege, N et al. (2011) Loss of T cell CD98 H chain specifically ablates T cell clonal expansion and protects from autoimmunity. The Journal of Immunology 187(2), 851860.Google Scholar
Carr, EL, Kelman, A, Wu, GS et al. (2010) Glutamine uptake and metabolism are coordinately regulated by ERK/MAPK during T lymphocyte activation. The Journal of Immunology 185(2), 10371044.Google Scholar
Carroll, B (2020) Spatial regulation of mTORC1 signalling: beyond the Rag GTPases. Seminars in Cell and Developmental Biology 107, 103111.Google Scholar
Castellano, F and Molinier-Frenkel, V (2020) Control of T-cell activation and signaling by amino-acid catabolizing enzymes. Frontiers in Cell and Developmental Biology 8, 613416.Google Scholar
Chaneton, B, Hillmann, P, Zheng, L et al. (2012) Serine is a natural ligand and allosteric activator of pyruvate kinase M2. Nature 491(7424), 458462.Google Scholar
Chen, S, Xia, Y, He, F et al. (2020) Serine supports IL-1β production in macrophages through mTOR signaling. Frontiers in Immunology 11, 1866.Google Scholar
Chen, Y, Xu, Z, Sun, H et al. (2023) Regulation of CD8+ T memory and exhaustion by the mTOR signals. Cellular and Molecular Immunology 20(9), 10231039.Google Scholar
Chi, H (2012) Regulation and function of mTOR signalling in T cell fate decisions. Nature Reviews Immunology 12(5), 325338.Google Scholar
Choudhury, FK (2021) Mitochondrial redox metabolism: the epicenter of metabolism during cancer progression. Antioxidants 10(11), 1838.Google Scholar
Ciofani, M and Zúñiga-Pflücker, JC (2010) Determining γδ versus αβ T cell development. Nature Reviews Immunology 10(9), 657663.Google Scholar
Clemens, MJ, Elia, A and Morley, SJ (2013) Requirement for the eIF4E binding proteins for the synergistic down-regulation of protein synthesis by hypertonic conditions and mTOR inhibition. PLOS ONE 8(8), e71138.Google Scholar
Collins, SM, Surette, M and Bercik, P (2012) The interplay between the intestinal microbiota and the brain. Nature Reviews, Microbiology 10(11), 735742.Google Scholar
Cui, Z, Joiner, AMN, Jansen, RM et al. (2023) Amino acid sensing and lysosomal signaling complexes. Current Opinion in Structural Biology 79, 102544.Google Scholar
Dai, W, Feng, J, Hu, X et al. (2021) SLC7A7 is a prognostic biomarker correlated with immune infiltrates in non-small cell lung cancer. Cancer Cell International 21(1), 106.Google Scholar
Dai, Z-L, Wu, G and Zhu, W-Y (2011) Amino acid metabolism in intestinal bacteria: links between gut ecology and host health. Frontiers in Bioscience 16(1), 17681786.Google Scholar
Dalmasso, G, Nguyen, HTT, Yan, Y et al. (2008) Butyrate transcriptionally enhances peptide transporter PepT1 expression and activity. PLoS One 3(6), e2476.Google Scholar
Darnell, AM, Subramaniam, AR and O’Shea, EK (2018) Translational control through differential ribosome pausing during amino acid limitation in mammalian cells. Molecular Cell 71(2), 229243.Google Scholar
Delmonte, OM, Villa, A and Notarangelo, LD (2020) Immune dysregulation in patients with RAG deficiency and other forms of combined immune deficiency. Blood 135(9), 610619.Google Scholar
Dimou, A, Tsimihodimos, V and Bairaktari, E (2022) The critical role of the branched chain amino acids (BCAAs) catabolism-regulating enzymes, branched-chain aminotransferase (BCAT) and branched-chain α-keto acid dehydrogenase (BCKD), in human pathophysiology. International Journal of Molecular Sciences 23(7), 4022.Google Scholar
Durek, P, Nordström, K, Gasparoni, G et al. (2016) Epigenomic profiling of human CD4+ T cells supports a linear differentiation model and highlights molecular regulators of memory development. Immunity 45(5), 11481161.Google Scholar
Edwards, DN, Ngwa, VM, Raybuck, AL et al. (2021) Selective glutamine metabolism inhibition in tumor cells improves antitumor T lymphocyte activity in triple-negative breast cancer. The Journal of Clinical Investigation 131(4), e140100.Google Scholar
Englander, MT, Avins, JL, Fleisher, RC et al. (2015) The ribosome can discriminate the chirality of amino acids within its peptidyl-transferase center. Proceedings of the National Academy of Sciences 112(19), 60386043.Google Scholar
Fanimo, A, Susenbeth, A and Südekum, K-H (2006) Protein utilisation, lysine bioavailability and nutrient digestibility of shrimp meal in growing pigs. Animal Feed Science and Technology 129(3-4), 196209.Google Scholar
Filardy, AA, Pires, DR, Nunes, MP et al. (2010) Proinflammatory clearance of apoptotic neutrophils induces an IL-12lowIL-10high regulatory phenotype in macrophage. The Journal of Immunology 185(4), 20442050.Google Scholar
Frye, M, Harada, BT, Behm, M et al. (2018) RNA modifications modulate gene expression during development. Science 361(6409), 13461349.Google Scholar
Fu, Y, Fu, Z, Su, Z et al. (2023) mLST8 is essential for coronavirus replication and regulates its replication through the mTORC1 pathway. Mbio 14(4), e0089900823.Google Scholar
Galloway, A, Kaskar, A, Ditsova, D et al. (2021) Upregulation of RNA cap methyltransferase RNMT drives ribosome biogenesis during T cell activation. Nucleic Acids Research 49(12), 67226738.Google Scholar
Gan, L, Seki, A, Shen, K et al. (2019) The lysosomal GPCR-like protein GPR137B regulates Rag and mTORC1 localization and activity. Nature Cell Biology 21(5), 614626.Google Scholar
Ge, T, Yang, J, Zhou, S et al. (2020) The role of the pentose phosphate pathway in diabetes and cancer. Frontiers in Endocrinology 11, 507678.Google Scholar
Ghoname, RM, Ghozlan, HA and Sabry, SAEF (2022) Novel research on Rapamycin analogue production by Streptomyces sp. strain RHS5. Journal of Advanced Biomedical and Pharmaceutical Sciences 5(2), 8895.Google Scholar
Groenewoud Marlous, J and Zwartkruis Fried, JT (2013) Rheb and Rags come together at the lysosome to activate mTORC1. Biochemical Society Transactions 41(4), 951955.Google Scholar
Grosso, S, Pesce, E, Brina, D et al. (2011) Sensitivity of global translation to mTOR inhibition in REN cells depends on the equilibrium between eIF4E and 4E-BP1. PLoS One 6(12), e29136.Google Scholar
Guha, I, Bhuniya, A, Shukla, D et al. (2020) Tumor arrests DN2 to DN3 Pro T cell transition and promotes its conversion to thymic dendritic cells by reciprocally regulating notch1 and ikaros signaling. Frontiers in Immunology 11, 898.Google Scholar
Gupta, S and Sarangi, PP (2023) Inflammation driven metabolic regulation and adaptation in macrophages. Clinical Immunology 246, 109216.Google Scholar
Han, C, Ge, M, Ho, P-C et al. (2021) Fueling T-cell antitumor immunity: amino acid metabolism revisited. Cancer Immunology Research 9(12), 13731382.Google Scholar
Hayashi, K, Jutabha, P, Endou, H et al. (2013) LAT1 is a critical transporter of essential amino acids for immune reactions in activated human T cells. The Journal of Immunology 191(8), 40804085.Google Scholar
He, F, Yin, Z, Wu, C et al. (2019) L-serine lowers the inflammatory responses during Pasteurella multocida infection. Infection and Immunity 87(12), 1128.Google Scholar
He, L, Liu, Y, Long, J et al. (2020) Maternal serine supply from late pregnancy to lactation improves offspring performance through modulation of metabolic pathways. Food & Function 11(9), 80898098.Google Scholar
He, Y, Liu, Y, Guan, P et al. (2023) Serine administration improves selenium status, oxidative stress, and mitochondrial function in longissimus dorsi muscle of piglets with intrauterine growth retardation. Biological Trace Element Research 201(4), 17401747.Google Scholar
Henning, AN, Roychoudhuri, R and Restifo, NP (2018) Epigenetic control of CD8+ T cell differentiation. Nature Reviews Immunology 18(5), 340356.Google Scholar
Hertel, A, Alves, LM, Dutz, H et al. (2022) USP32-regulated LAMTOR1 ubiquitination impacts mTORC1 activation and autophagy induction. Cell Reports 41(10), 111653.Google Scholar
Hoebeke, I, De Smedt, M, Stolz, F et al. (2007) T-, B- and NK-lymphoid, but not myeloid cells arise from human CD34+CD38CD7+ common lymphoid progenitors expressing lymphoid-specific genes. Leukemia 21(2), 311319.Google Scholar
Hope, HC and Salmond, RJ (2021) The role of non-essential amino acids in T cell function and anti-tumour immunity. Archivum Immunologiae Et Therapiae Experimentalis 69(1), 29.Google Scholar
Huang, H, Long, L, Zhou, P et al. (2020) mTOR signaling at the crossroads of environmental signals and T-cell fate decisions. Immunological Reviews 295(1), 1538.Google Scholar
Huang, R, Wang, H, Hong, J et al. (2023) Targeting glutamine metabolic reprogramming of SLC7A5 enhances the efficacy of anti-PD-1 in triple-negative breast cancer. Frontiers in Immunology 14, 1251643.Google Scholar
Jensen, H, Potempa, M, Gotthardt, D et al. (2017) Cutting edge: IL-2-induced expression of the amino acid transporters SLC1A5 and CD98 is a prerequisite for NKG2D-mediated activation of human NK cells. The Journal of Immunology 199(6), 19671972.Google Scholar
Jha, AK, Huang, S-C-C, Sergushichev, A et al. (2015) Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Immunity 42(3), 419430.Google Scholar
Jiang, C, Dai, X, He, S et al. (2023) Ring domains are essential for GATOR2-dependent mTORC1 activation. Molecular Cell 83(1), 7489.Google Scholar
Jiang, S, Yan, W, Wang, SE et al. (2018) Let-7 suppresses B cell activation through restricting the availability of necessary nutrients. Cell Metabolism 27(2), 393403.Google Scholar
Johnson, MO, Wolf, MM, Madden, MZ et al. (2018) Distinct regulation of Th17 and Th1 cell differentiation by glutaminase-dependent metabolism. Cell 175(7), 1780–1795.e1719.Google Scholar
Ju, W, Yun, M, Jang, Y et al. (2008) Comparison of synthetic lysine sources on growth performance, nutrient digestibility and nitrogen retention in weaning pigs. Asian-Australasian Journal of Animal Sciences 21(1), 9096.Google Scholar
Kaech, SM and Cui, W (2012) Transcriptional control of effector and memory CD8+ T cell differentiation. Nature Reviews Immunology 12(11), 749761.Google Scholar
Kaech, SM, Hemby, S, Kersh, E et al. (2002) Molecular and functional profiling of memory CD8 T cell differentiation. Cell 111(6), 837851.Google Scholar
Kan, MJ, Lee, JE, Wilson, JG et al. (2015) Arginine deprivation and immune suppression in a mouse model of Alzheimer’s disease. Journal of Neuroscience 35(15), 59695982.Google Scholar
Kang, YJ, Song, W, Lee, SJ et al. (2024) Inhibition of BCAT1-mediated cytosolic leucine metabolism regulates Th17 responses via the mTORC1-HIF1α pathway. Experimental & Molecular Medicine 56(8), 17761790.Google Scholar
Karsunky, H, Inlay, MA, Serwold, T et al. (2008) Flk2+ common lymphoid progenitors possess equivalent differentiation potential for the B and T lineages. Blood 111(12), 55625570.Google Scholar
Kedersha, N, Chen, S, Gilks, N et al. (2002) Evidence that ternary complex (eIF2-GTP-tRNAi Met)–deficient preinitiation complexes are core constituents of mammalian stress granules. Molecular Biology of the Cell 13(1), 195210.Google Scholar
Kedl, RM, Rees, WA, Hildeman, DA et al. (2000) T Cells compete for access to antigen-bearing antigen-presenting cells. Journal of Experimental Medicine 192(8), 11051114.Google Scholar
Kelly, B and Pearce, EL (2020) Amino assets: how amino acids support immunity. Cell Metabolism 32(2), 154175.Google Scholar
Kempkes, RWM, Joosten, I, Koenen, HJPM et al. (2019) Metabolic pathways involved in regulatory T cell functionality. Frontiers in Immunology 10, 483290.Google Scholar
Kim, J and Kim, E (2016) Rag GTPase in amino acid signaling. Amino Acids 48(4), 915928.Google Scholar
Kim, Y, Sundrud, MS, Zhou, C et al. (2020) Aminoacyl-tRNA synthetase inhibition activates a pathway that branches from the canonical amino acid response in mammalian cells. Proceedings of the National Academy of Sciences 117(16), 89008911.Google Scholar
Kitada, M, Xu, J, Ogura, Y et al. (2020) Mechanism of activation of mechanistic target of rapamycin complex 1 by methionine. Frontiers in Cell and Developmental Biology 8, 715.Google Scholar
Kolliniati, O, Ieronymaki, E, Vergadi, E et al. (2021) Metabolic regulation of macrophage activation. Journal of Innate Immunity 14(1), 5168.Google Scholar
Kono, M, Yoshida, N, Maeda, K et al. (2018) Transcriptional factor ICER promotes glutaminolysis and the generation of Th17 cells. Proceedings of the National Academy of Sciences 115(10), 24782483.Google Scholar
Lama-Sherpa, TD, Jeong, M-H and Jewell, JL (2023) Regulation of mTORC1 by the Rag GTPases. Biochemical Society Transactions 51(2), 655664.Google Scholar
Lane, AN and Fan, TW (2015) Regulation of mammalian nucleotide metabolism and biosynthesis. Nucleic Acids Research 43(4), 24662485.Google Scholar
Lees, JG, Gardner, DK and Harvey, AJ (2017) Pluripotent stem cell metabolism and mitochondria: beyond ATP. Stem Cells International 2017(1), 2874283.Google Scholar
Le Page, L, Baldwin, CL and Telfer, JC (2022) γδ T cells in artiodactyls: focus on swine. Developmental & Comparative Immunology 128, 104334.Google Scholar
Li, H-B, Tong, J, Zhu, S et al. (2017) m6A mRNA methylation controls T cell homeostasis by targeting the IL-7/STAT5/SOCS pathways. Nature 548(7667), 338342.Google Scholar
Li, J, Yin, L, Wang, L et al. (2019) Effects of vitamin B6 on growth, diarrhea rate, intestinal morphology, function, and inflammatory factors expression in a high-protein diet fed to weaned piglets. Journal of Animal Science 97(12), 48654874.Google Scholar
Li, MO, Wan, YY and Flavell, RA (2007a) T cell-produced transforming growth factor-β1 controls T cell tolerance and regulates Th1-and Th17-cell differentiation. Immunity 26(5), 579591.Google Scholar
Li, P, Spolski, R, Liao, W et al. (2014) Complex interactions of transcription factors in mediating cytokine biology in T cells. Immunological Reviews 261(1), 141156.Google Scholar
Li, P, Yin, Y-L, Li, D et al. (2007b) Amino acids and immune function. British Journal of Nutrition 98(2), 237252.Google Scholar
Li, S, Wu, Q, Jiang, Z et al. (2022) miR-31-5p regulates type I interferon by targeting SLC15A4 in Plasmacytoid dendritic cells of systemic lupus erythematosus. Journal of Inflammation Research 15, 66076616.Google Scholar
Li, W and Zhang, L (2020) Rewiring mitochondrial metabolism for CD8+ T cell memory formation and effective cancer immunotherapy. Frontiers in Immunology 11, 526982.Google Scholar
Liao, Y, Fan, L, Bin, P et al. (2022) GABA signaling enforces intestinal germinal center B cell differentiation. Proceedings of the National Academy of Sciences 119(44), e2215921119.Google Scholar
Licence, S and Binns, R (1995) Major long-term changes in gamma delta T-cell receptor-positive and CD2+ T-cell subsets after neonatal thymectomy in the pig: a longitudinal study lasting nearly 2 years. Immunology 85(2), 276.Google Scholar
Lin, L, Ren, R, Xiong, Q et al. (2024) Remodeling of T-cell mitochondrial metabolism to treat autoimmune diseases. Autoimmunity Reviews 23(6), 103583.Google Scholar
Liu, A, Wang, Y, Ding, Y et al. (2015) Cutting edge: Hematopoietic stem cell expansion and common lymphoid progenitor depletion require hematopoietic-derived, cell-autonomous TLR4 in a model of chronic endotoxin. The Journal of Immunology 195(6), 25242528.Google Scholar
Liu, H-P, Cao, AT, Feng, T et al. (2015) TGF-β converts Th1 cells into Th17 cells through stimulation of Runx1 expression. European Journal of Immunology 45(4), 10101018.Google Scholar
Liu, P-S, Wang, H, Li, X et al. (2017) α-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nature Immunology 18(9), 985994.Google Scholar
Liu, Z, Le, Y, Chen, H et al. (2021) Role of PKM2-mediated immunometabolic reprogramming on development of cytokine storm. Frontiers in Immunology 12, 748573.Google Scholar
Loftus, RM, Assmann, N, Kedia-Mehta, N et al. (2018) Amino acid-dependent cMyc expression is essential for NK cell metabolic and functional responses in mice. Nature Communications 9(1), 2341.Google Scholar
Ma, EH, Bantug, G, Griss, T et al. (2017) Serine is an essential metabolite for effector T cell expansion. Cell Metabolism 25(2), 345357.Google Scholar
Ma, G, Zhang, Z, Li, P et al. (2022) Reprogramming of glutamine metabolism and its impact on immune response in the tumor microenvironment. Cell Communication and Signaling 20(1), 114.Google Scholar
Ma, N, Guo, P, Zhang, J et al. (2018) Nutrients mediate intestinal bacteria–mucosal immune crosstalk. Frontiers in Immunology 9, 5.Google Scholar
Ma, S, Ming, Y, Wu, J et al. (2024) Cellular metabolism regulates the differentiation and function of T-cell subsets. Cellular and Molecular Immunology 21(5), 419435.Google Scholar
Macfarlane, GT and Macfarlane, S (2012) Bacteria, colonic fermentation, and gastrointestinal health. Journal of AOAC International 95(1), 5060.Google Scholar
Maloy, KJ, Salaun, L, Cahill, R et al. (2003) CD4+ CD25+ TR cells suppress innate immune pathology through cytokine-dependent mechanisms. The Journal of Experimental Medicine 197(1), 111119.Google Scholar
Marchingo, JM, Sinclair, LV, Howden, AJM et al. (2020) Quantitative analysis of how Myc controls T cell proteomes and metabolic pathways during T cell activation. eLife 9, e53725.Google Scholar
Marelli-Berg, FM, Fu, H and Mauro, C (2012) Molecular mechanisms of metabolic reprogramming in proliferating cells: Implications for T-cell-mediated immunity. Immunology 136(4), 363369.Google Scholar
Marie, JC, Letterio, JJ, Gavin, M et al. (2005) TGF-β1 maintains suppressor function and Foxp3 expression in CD4+ CD25+ regulatory T cells. The Journal of Experimental Medicine 201(7), 10611067.Google Scholar
Martíi Líndez, -A-A and Reith, W (2021) Arginine-dependent immune responses. Cellular and Molecular Life Sciences 78(13), 53035324.Google Scholar
Michalek, RD, Gerriets, VA, Jacobs, SR et al. (2011) Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. The Journal of Immunology 186(6), 32993303.Google Scholar
Minton, DR, Nam, M, McLaughlin, DJ et al. (2018) Serine catabolism by SHMT2 is required for proper mitochondrial translation initiation and maintenance of formylmethionyl-tRNAs. Molecular Cell 69(4), 610621.Google Scholar
Montacchiesi, G and Pace, L (2022) Epigenetics and CD8+ T cell memory. Immunological Reviews 305(1), 7789.Google Scholar
Montoya, M, Schiavoni, G, Mattei, F et al. (2002) Type I interferons produced by dendritic cells promote their phenotypic and functional activation. Blood 99(9), 32633271.Google Scholar
Mota-Martorell, N, Jove, M, Pradas, I et al. (2020) Gene expression and regulatory factors of the mechanistic target of rapamycin (mTOR) complex 1 predict mammalian longevity. Geroscience 42(4), 11571173.Google Scholar
Muri, J and Kopf, M (2021) Redox regulation of immunometabolism. Nature Reviews Immunology 21(6), 363381.Google Scholar
Myrie, SB, Bertolo, RF, Sauer, WC et al. (2008) Effect of common antinutritive factors and fibrous feedstuffs in pig diets on amino acid digestibilities with special emphasis on threonine. Journal of Animal Science 86(3), 609619.Google Scholar
Neinast, MD, Jang, C, Hui, S et al. (2019) Quantitative analysis of the whole-body metabolic fate of branched-chain amino acids. Cell Metabolism 29(2), 417429.Google Scholar
Nguyen, DC, Duan, M, Ali, M et al. (2021) Plasma cell survival: the intrinsic drivers, migratory signals, and extrinsic regulators. Immunological Reviews 303(1), 138153.Google Scholar
Nguyen, TP, Frank, AR and Jewell, JL (2017) Amino acid and small GTPase regulation of mTORC1. Cellular Logistics 7(4), e1378794.Google Scholar
Oh, M-H, Sun, I-H, Zhao, L et al. (2020) Targeting glutamine metabolism enhances tumor-specific immunity by modulating suppressive myeloid cells. The Journal of Clinical Investigation 130(7), 38653884.Google Scholar
Ohh, S, Han, K, Chae, B et al. (2002) Effects of feed processing methods on growth performance and ileal digestibility of amino acids in young pigs. Asian-Australasian Journal of Animal Sciences 15(12), 17651772.Google Scholar
O’Shea, JJ, Steward-Tharp, SM, Laurence, A et al. (2009) Signal transduction and Th17 cell differentiation. Microbes and Infection 11(5), 599611.Google Scholar
Palmieri, EM, Gonzalez-Cotto, M, Baseler, WA et al. (2020) Nitric oxide orchestrates metabolic rewiring in M1 macrophages by targeting aconitase 2 and pyruvate dehydrogenase. Nature Communications 11(1), 698.Google Scholar
Palsson-mcdermott, EM, Curtis Anne, M, Goel, G et al. (2015) Pyruvate kinase M2 regulates Hif-1α activity and IL-1β induction and is a critical determinant of the Warburg effect in LPS-activated macrophages. Cell Metabolism 21(2), 347.Google Scholar
Papathanassiu, AE, Ko, J-H, Imprialou, M et al. (2017) BCAT1 controls metabolic reprogramming in activated human macrophages and is associated with inflammatory diseases. Nature Communications 8(1), 16040.Google Scholar
Patsoukis, N, Bardhan, K, Weaver, J et al. (2016) The role of metabolic reprogramming in T cell fate and function. Current Trends in Immunology 17, 112.Google Scholar
Pearce, EL and Pearce, EJ (2013) Metabolic pathways in immune cell activation and quiescence. Immunity 38(4), 633643.Google Scholar
Pearce, EL, Walsh, MC, Cejas, PJ et al. (2009) Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature 460(7251), 103107.Google Scholar
Pelechano, V and Alepuz, P (2017) eIF5A facilitates translation termination globally and promotes the elongation of many non polyproline-specific tripeptide sequences. Nucleic Acids Research 45(12), 73267338.Google Scholar
Peng, X, Hu, L, Liu, Y et al. (2016) Effects of low-protein diets supplemented with indispensable amino acids on growth performance, intestinal morphology and immunological parameters in 13 to 35 kg pigs. Animal 10(11), 18121820.Google Scholar
Pollizzi, KN, Sun, I-H, Patel, CH et al. (2016) Asymmetric inheritance of mTORC1 kinase activity during division dictates CD8+ T cell differentiation. Nature Immunology 17(6), 704711.Google Scholar
Puckett, DL, Alquraishi, M, Chowanadisai, W et al. (2021) The role of PKM2 in metabolic reprogramming: insights into the regulatory roles of non-coding RNAs. International Journal of Molecular Sciences 22(3), 1171.Google Scholar
Puleston, DJ, Buck, MD, Klein Geltink, RI et al. (2019) Polyamines and eIF5A hypusination modulate mitochondrial respiration and macrophage activation. Cell Metabolism 30(2), 352363.Google Scholar
Quintin, J, Saeed, S, Martens Joost, HA et al. (2012) Candida albicans infection affords protection against reinfection via functional reprogramming of monocytes. Cell Host & Microbe 12(2), 223232.Google Scholar
Rashidi, A, Miska, J, Lee-Chang, C et al. (2020) GCN2 is essential for CD8+ T cell survival and function in murine models of malignant glioma. Cancer Immunology, Immunotherapy 69(1), 8194.Google Scholar
Ren, W, Liu, G, Chen, S et al. (2017a) Melatonin signaling in T cells: functions and applications. Journal of Pineal Research 62(3), e12394.Google Scholar
Ren, W, Rajendran, R, Zhao, Y et al. (2018) Amino acids as mediators of metabolic cross talk between host and pathogen. Frontiers in Immunology 9, 319.Google Scholar
Ren, W, Yin, J, Xiao, H et al. (2017b) Intestinal microbiota-derived GABA mediates interleukin-17 expression during enterotoxigenic Escherichia coli infection. Frontiers in Immunology 7, 685.Google Scholar
Robinson, MJ, Dowling, MR, Pitt, C et al. (2022) Long-lived plasma cells accumulate in the bone marrow at a constant rate from early in an immune response. Science Immunology 7(76), eabm8389.Google Scholar
Ron-Harel, N, Ghergurovich, JM, Notarangelo, G et al. (2019) T cell activation depends on extracellular alanine. Cell Reports 28(12), 30113021.Google Scholar
Roy, DG, Chen, J, Mamane, V et al. (2020) Methionine metabolism shapes T helper cell responses through regulation of epigenetic reprogramming. Cell Metabolism 31(2), 250266.Google Scholar
, AGA, Moreno, YMF and Carciofi, BAM (2020) Food processing for the improvement of plant proteins digestibility. Critical Reviews in Food Science and Nutrition 60(20), 33673386.Google Scholar
Saba, I, Kosan, C, Vassen, L et al. (2011) IL-7R–dependent survival and differentiation of early T-lineage progenitors is regulated by the BTB/POZ domain transcription factor Miz-1. Blood 117(12), 33703381.Google Scholar
Sancak, Y, Bar-Peled, L, Zoncu, R et al. (2010) Ragulator-Rag complex targets mTORC1 to the lysosomal surface and is necessary for its activation by amino acids. Cell 141(2), 290303.Google Scholar
Sancak, Y, Peterson, TR, Shaul, YD et al. (2008) The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1. Science 320(5882), 14961501.Google Scholar
Schalk, C, Pfaffinger, B, Schmucker, S et al. (2019) Pregnancy-associated alterations of peripheral blood immune cell numbers in domestic sows are modified by social rank. Animals 9(3), 112.Google Scholar
Schmidl, C, Delacher, M, Huehn, J et al. (2018) Epigenetic mechanisms regulating T-cell responses. Journal of Allergy and Clinical Immunology 142(3), 728743.Google Scholar
Schuller, AP, Wu, CC, Dever, TE et al. (2017) eIF5A functions globally in translation elongation and termination. Molecular Cell 66(2), 194205.Google Scholar
Shan, X, Hu, P, Ni, L et al. (2022) Serine metabolism orchestrates macrophage polarization by regulating the IGF1-p38 axis. Cellular and Molecular Immunology 19(11), 12631278.Google Scholar
Shen, K, Valenstein, ML, Gu, X et al. (2019) Arg-78 of Nprl2 catalyzes GATOR1-stimulated GTP hydrolysis by the Rag GTPases. Journal of Biological Chemistry 294(8), 29705944.Google Scholar
Shi, H, Chapman, NM, Wen, J et al. (2019) Amino acids license kinase mTORC1 activity and Treg cell function via small G proteins Rag and Rheb. Immunity, 51(6), 10121027.Google Scholar
Shi, LZ, Wang, R, Huang, G et al. (2011) HIF1α–dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. Journal of Experimental Medicine 208(7), 13671376.Google Scholar
Shirai, T, Nazarewicz, RR, Wallis, BB et al. (2016) The glycolytic enzyme PKM2 bridges metabolic and inflammatory dysfunction in coronary artery disease. Journal of Experimental Medicine 213(3), 337354.Google Scholar
Sinclair, LV, Howden, AJM, Brenes, A et al. (2019) Antigen receptor control of methionine metabolism in T cells. eLife 8, e44210.Google Scholar
Sinclair, LV, Rolf, J, Emslie, E et al. (2013) Control of amino-acid transport by antigen receptors coordinates the metabolic reprogramming essential for T cell differentiation. Nature Immunology 14(5), 500508.Google Scholar
Song, W, Li, D, Tao, L et al. (2020) Solute carrier transporters: The metabolic gatekeepers of immune cells. Acta PharmaceuticaSinica B, 10(1), 6178.Google Scholar
Sonner, JK, Deumelandt, K, Ott, M et al. (2016) The stress kinase GCN2 does not mediate suppression of antitumor T cell responses by tryptophan catabolism in experimental melanomas. OncoImmunology 5(12), e1240858.Google Scholar
Stone, TW and Williams, RO (2023) Modulation of T cells by tryptophan metabolites in the kynurenine pathway. Trends in Pharmacological Sciences 44(7), 442456.Google Scholar
Sun, L, Fu, J and Zhou, Y (2017) Metabolism controls the balance of Th17/T-regulatory cells. Frontiers in Immunology 8, 1632.Google Scholar
Tabas, I and Bornfeldt, KE (2020) Intracellular and intercellular aspects of macrophage immunometabolism in atherosclerosis. Circulation Research 126(9), 12091227.Google Scholar
Tae, K, Kim, S-J, S.-w., C et al. (2023) L-type amino acid transporter 1 (LAT1) promotes PMA-Induced cell migration through mTORC2 activation at the lysosome. Cells 12(20), 2504.Google Scholar
Tang, Z, Wang, Y, Xing, R et al. (2020) Deltex-1 is indispensible for the IL-6 and TGF-β treatment-triggered differentiation of Th17 cells. Cellular Immunology 356, 104176.Google Scholar
Tani, H, Ohnishi, S, Shitara, H et al. (2018) Mice deficient in the Shmt2 gene have mitochondrial respiration defects and are embryonic lethal. Scientific Reports 8(1), 425.Google Scholar
Tantawy, AA and Naguib, DM (2019) Arginine, histidine and tryptophan: a new hope for cancer immunotherapy. PharmaNutrition 8, 100149.Google Scholar
Thangavelu, G, Andrejeva, G, Bolivar-Wagers, S et al. (2022) Retinoic acid signaling acts as a rheostat to balance Treg function. Cellular and Molecular Immunology 19(7), 820833.Google Scholar
Tomé, D (2021) Amino acid metabolism and signalling pathways: potential targets in the control of infection and immunity. Nutrition and Diabetes 11(1), 20.Google Scholar
Traba, J, Sack, MN, Waldmann, TA et al. (2021) Immunometabolism at the nexus of cancer therapeutic Efficacy and resistance. Frontiers in Immunology 12, 657293.Google Scholar
Tsujimoto, K, Takamatsu, H and Kumanogoh, A (2023) The Ragulator complex: Delving its multifunctional impact on metabolism and beyond. Inflammation and Regeneration 43(1), 28.Google Scholar
Valenstein, ML, Lalgudi, PV, Gu, X et al. (2024) Rag–Ragulator is the central organizer of the physical architecture of the mTORC1 nutrient-sensing pathway. Proceedings of the National Academy of Sciences 121(35), e2322755121.Google Scholar
Verbist, KC, Guy, CS, Milasta, S et al. (2016) Metabolic maintenance of cell asymmetry following division in activated T lymphocytes. Nature 532(7599), 389393.Google Scholar
Waickman, AT and Powell, JD (2012) mTOR, metabolism, and the regulation of T-cell differentiation and function. Immunological Reviews 249(1), 4358.Google Scholar
Wang, C, Chen, Q, Chen, S et al. (2024) Serine synthesis sustains macrophage IL-1β production via NAD+-dependent protein acetylation. Molecular Cell 84(4), 744759.Google Scholar
Wang, C, Peng, Y, Zhang, Y et al. (2023) The biological functions and metabolic pathways of valine in swine. Journal of Animal Science and Biotechnology 14(1), 135.Google Scholar
Wang, D, Wan, X, Du, X et al. (2021a) Insights into the Interaction of Lysosomal Amino Acid Transporters SLC38A9 and SLC36A1 Involved in mTORC1 Signaling in C2C12 Cells. Biomolecules 11(9), 1314.Google Scholar
Wang, F, Qi, Z, Yao, Y et al. (2021b) Exploring the stage-specific roles of Tcf-1 in T cell development and malignancy at single-cell resolution. Cellular and Molecular Immunology 18(3), 644659.Google Scholar
Wang, K, Zhao, W, Jin, R et al. (2021c) Thymic iNKT cell differentiation at single-cell resolution. Cellular and Molecular Immunology 18(8), 20652066.Google Scholar
Wang, S, Schianchi, F, Neumann, D et al. (2021d) Specific amino acid supplementation rescues the heart from lipid overload-induced insulin resistance and contractile dysfunction by targeting the endosomal mTOR–v-ATPase axis. Molecular Metabolism 53, 101293.Google Scholar
Wang, W and Zou, W (2020) Amino acids and their transporters in T cell immunity and cancer therapy. Molecular Cell 80(3), 384395.Google Scholar
Wang, Y, Lei, J, Zhang, S et al. (2022) 4EBP1 senses extracellular glucose deprivation and initiates cell death signaling in lung cancer. Cell Death and Disease 13(12), 1075.Google Scholar
Wang, Z, Guan, D, Wang, S et al. (2020) Glycolysis and oxidative phosphorylation play critical roles in natural killer cell receptor-mediated natural killer cell functions. Frontiers in Immunology 11, 202.Google Scholar
Wiarda, JE, Trachsel, JM, Bond, ZF et al. (2020) Intraepithelial T cells diverge by intestinal location as pigs age. Frontiers in Immunology 11, 540164.Google Scholar
Winter, SJ and Krueger, A (2019) Development of unconventional T cells controlled by microRNA. Frontiers in Immunology 10, 489268.Google Scholar
Woyengo, T and Nyachoti, C (2013) Anti-nutritional effects of phytic acid in diets for pigs and poultry–current knowledge and directions for future research. Canadian Journal of Animal Science 93(1), 921.Google Scholar
Wu, -C-C-C, Zinshteyn, B, Wehner, KA et al. (2019) High-resolution ribosome profiling defines discrete ribosome elongation states and translational regulation during cellular stress. Molecular Cell 73(5), 959970.Google Scholar
Wu, G, Bazer, FW, Johnson, GA et al. (2018) Board-invited review: arginine nutrition and metabolism in growing, gestating, and lactating swine. Journal of Animal Science 96(12), 50355051.Google Scholar
Xia, Y, Chen, S, Zeng, S et al. (2019) Melatonin in macrophage biology: current understanding and future perspectives. Journal of Pineal Research 66(2), e12547.Google Scholar
Xie, M, Yu, Y, Kang, R et al. (2016) PKM2-dependent glycolysis promotes NLRP3 and AIM2 inflammasome activation. Nature Communications 7(1), 13280.Google Scholar
Xu, A, Bhanumathy, KK, Wu, J et al. (2016) IL-15 signaling promotes adoptive effector T-cell survival and memory formation in irradiation-induced lymphopenia. Cell and Bioscience 6(1), 30.Google Scholar
Xuekai, L, Yan, S, Jian, C et al. (2024) Advances in reprogramming of energy metabolism in tumor T cells. Frontiers in Immunology 15, 1347181.Google Scholar
Yan, Y, Zhang, G-X, Gran, B et al. (2010) IDO upregulates regulatory T cells via tryptophan catabolite and suppresses encephalitogenic T cell responses in experimental autoimmune encephalomyelitis. The Journal of Immunology 185(10), 59535961.Google Scholar
Yang, L, Chu, Z, Liu, M et al. (2023) Amino acid metabolism in immune cells: essential regulators of the effector functions, and promising opportunities to enhance cancer immunotherapy. Journal of Hematology & Oncology 16(1), 59.Google Scholar
Yang, M, Lu, Y, Piao, W et al. (2022) The translational regulation in mTOR pathway. Biomolecules 12(6), 802.Google Scholar
Yang, Z and Liao, SF (2019) Physiological effects of dietary amino acids on gut health and functions of swine. Frontiers in Veterinary Science 6, 169.Google Scholar
Yaqoob, P and Calder, PC (1997) Glutamine requirement of proliferating T lymphocytes. Nutrition 13(7-8), 646651.Google Scholar
Yeramian, A, Martin, L, Serrat, N et al. (2006) Arginine transport via cationic amino acid transporter 2 plays a critical regulatory role in classical or alternative activation of macrophages. The Journal of Immunology 176(10), 59185924.Google Scholar
Yoon, BR, Oh, Y-J, Kang, SW et al. (2018) Role of SLC7A5 in metabolic reprogramming of human monocyte/macrophage immune responses. Frontiers in Immunology 9, 53.Google Scholar
Yu, Q, Tu, H, Yin, X et al. (2022) Targeting glutamine metabolism ameliorates autoimmune hepatitis via inhibiting T cell activation and differentiation. Frontiers in Immunology 13, 880262.Google Scholar
Yui, MA, Feng, N and Rothenberg, EV (2010) Fine-scale staging of T cell lineage commitment in adult mouse thymus. The Journal of Immunology 185(1), 284293.Google Scholar
Zelante, T, Iannitti, RG, Cunha, C et al. (2013) Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 39(2), 372385.Google Scholar
Zhang, C, Wang, H, Yin, L et al. (2020) Immunometabolism in the pathogenesis of systemic lupus erythematosus. Journal of Translational Autoimmunity 3, 100046.Google Scholar
Zhang, H, Hua, R, Zhang, B et al. (2018) Serine alleviates dextran sulfate sodium-induced colitis and regulates the gut microbiota in mice. Frontiers in Microbiology 9, 3062.Google Scholar
Zhang, S, Gang, X, Yang, S et al. (2021a) The alterations in and the role of the Th17/Treg balance in metabolic diseases. Frontiers in Immunology 12, 678355.Google Scholar
Zhang, S, Zhang, J, Yu, J et al. (2021b) Hyperforin ameliorates imiquimod-induced psoriasis-like murine skin inflammation by modulating IL-17A-producing γδ T cells. Frontiers in Immunology 12, 635076.Google Scholar
Zhao, H, Raines, LN and Huang, SC (2020a) Carbohydrate and amino acid metabolism as hallmarks for innate immune cell activation and function. Cells 9(3), 562.Google Scholar
Zhao, M, Wang, DD-H, Liu, X et al. (2020b) Metabolic modulation of macrophage function post myocardial infarction. Frontiers in Physiology 11, 674.Google Scholar
Zhao, T, Guan, Y, Xu, C et al. (2023) VWCE modulates amino acid-dependent mTOR signaling and coordinates with KICSTOR to recruit GATOR1 to the lysosomes. Nature Communications 14(1), 8464.Google Scholar
Zheng, X, Zhu, Y, Zhao, Z et al. (2023a) The role of amino acid metabolism in inflammatory bowel disease and other inflammatory diseases. Frontiers in Immunology 14, 284133.Google Scholar
Zheng, Y, Yao, Y, Ge, T et al. (2023b) Amino acid metabolism reprogramming: shedding new light on T cell anti-tumor immunity. Journal of Experimental & Clinical Cancer Research 42(1), 291.Google Scholar
Zhou, X, Zhang, Y, He, L et al. (2017) Serine prevents LPS-induced intestinal inflammation and barrier damage via p53-dependent glutathione synthesis and AMPK activation. Journal of Functional Foods 39, 225232.Google Scholar
Zhu, M and Wang, X (2020) Regulation of mTORC1 by small GTPases in response to nutrients. The Journal of Nutrition 150(5), 10041011.Google Scholar
Zhu, X, Wu, Y, Li, Y et al. (2024) The nutrient-sensing Rag-GTPase complex in B cells controls humoral immunity via TFEB/TFE3-dependent mitochondrial fitness. Nature Communications 15(1), 10163.Google Scholar
Figure 0

Figure 1. Mechanism of amino acid sensing by immune cells. When amino acid levels are low, uncharged tRNA activates GCN2, which interferes with the recruitment of mTORC1 substrates and blocks protein synthesis. In contrast, when amino acid levels are sufficient, cytoplasmic amino acid sensors inhibit GATOR1 through GATOR2, ultimately activating mTORC1 and promoting protein synthesis.

Figure 1

Figure 2. Development of T cells. T-cell development proceeds through a series of stages. DN1 cells can differentiate into B cells, myeloid cells, and innate T cells, while DN2b and DN3a cells can give rise to γδ T cells. At the DN3 stage, the pre-TCR complex-formed by TCRβ, pTα, and CD3 molecules-promotes β selection and drives the transition from DN3 to DN4. Both the pre-TCR and Notch signals are crucial for β selection and for the shift from the DN to the DP stage. After positive selection in the thymic cortex and negative selection in the thymic medulla, DP cells eventually differentiate into CD4+ T cells, CD8+ T cells, or iNKT cells.

Figure 2

Figure 3. Amino acid metabolism in activated T cell. Serine is required for the production of cytokines in activated T cells with the help of the key glycolytic enzyme PKM2. Although pyruvate can be used to make alanine, activated T cells reduce the synthesis of alanine from pyruvate in order to conserve pyruvate metabolism and convert it into acetyl-CoA for TCA cycle activity. Branched-chain amino acids (BCAAs) provide the TCA cycle with the intermediate product CoA. Glutamine and leucine also contribute to the TCA cycle via glutamate to α-ketoglutarate.

Figure 3

Figure 4. Sulfur-containing amino acids maintain redox status homeostasis in T cells. In T cells, methionine can be converted into cysteine. During this process, the intermediate S-adenosylmethionine donates methyl groups that modify immune effector proteins and nucleic acids, promoting cytokine gene expression in T cells and supporting innate and adaptive immune memory. Serine can also be transformed into cysteine, which contributes to the synthesis of the antioxidant glutathione and the formation of iron-sulfur (FeS) clusters.