Next Article in Journal
Correction: Zanin et al. Analysis of Radiation Toxicity in Mammalian Cells Stably Transduced with Mitochondrial Stat3. Int. J. Mol. Sci. 2023, 24, 8232
Previous Article in Journal
Different Dosages of Progesterone in Luteal Phase Support Reflect Varying Endometrial microRNA Expression in Frozen Embryo Transfer Cycles
Previous Article in Special Issue
IRF1 Mediates Growth Arrest and the Induction of a Secretory Phenotype in Alveolar Epithelial Cells in Response to Inflammatory Cytokines IFNγ/TNFα
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The PD-1/PD-L1 Axis in the Biology of MASLD

by
Rosaria Maria Pipitone
*,†,
Giulia Lupo
,
Rossella Zito
,
Ayesha Javed
,
Salvatore Petta
,
Grazia Pennisi
and
Stefania Grimaudo
*,‡
Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Piazza delle Cliniche, 90127 Palermo, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors also contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(7), 3671; https://doi.org/10.3390/ijms25073671
Submission received: 15 January 2024 / Revised: 18 March 2024 / Accepted: 19 March 2024 / Published: 25 March 2024
(This article belongs to the Collection Immunopathology and Immunosenescence)

Abstract

:
Metabolic Dysfunction-Associated Steatotic Liver (MASL), previously named nonalcoholic fatty liver (NAFL), is a multifactorial disease in which metabolic, genetic, and environmental risk factors play a predominant role. Obesity and type 2 diabetes act as triggers of the inflammatory response, which contributes to the progression of MASL to Metabolic Dysfunction-Associated Steatohepatitis and the development of hepatocellular carcinoma. In the liver, several parenchymal, nonparenchymal, and immune cells maintain immunological homeostasis, and different regulatory pathways balance the activation of the innate and adaptative immune system. PD-1/PD-L1 signaling acts, in the maintenance of the balance between the immune responses and the tissue immune homeostasis, promoting self-tolerance through the modulation of activated T cells. Recently, PD-1 has received much attention for its roles in inducing an exhausted T cells phenotype, promoting the tumor escape from immune responses. Indeed, in MASLD, the excessive fat accumulation dysregulates the immune system, increasing cytotoxic lymphocytes and decreasing their cytolytic activity. In this context, T cells exacerbate liver damage and promote tumor progression. The aim of this review is to illustrate the main pathogenetic mechanisms by which the immune system promotes the progression of MASLD and the transition to HCC, as well as to discuss the possible therapeutic applications of PD-1/PD-L1 target therapy to activate T cells and reinvigorate immune surveillance against cancer.

1. Introduction

Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD), previously named Nonalcoholic Fatty Liver Disease (NAFLD), is the most chronic liver disease due to the associated complex metabolic disorders and the high risk of complications, such as liver decompensation, hepatocellular carcinoma (HCC), and cardiovascular diseases [1,2,3]. Indeed, until a few years ago, NAFLD was considered the hepatic manifestation of Metabolic Syndrome (MetS) in terms of insulin resistance (IR), atherogenic dyslipidemia, abdominal obesity, and hypertension [4,5]. Recently, its terminology has been revised and updated, considering NAFLD a key factor in MeS [6], associated with metabolic disorders, such as obesity and type 2 diabetes (T2D) [5,7]. All these aspects led to revising the definition of NAFLD and considering it a metabolic disease. For this reason, it has recently been renamed Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) and Metabolic Dysfunction-Associated Steatohepatitis (MASH). In addition, the coexistence of metabolic and alcoholic liver disease has been identified, and the requirement of the absence of excessive alcohol consumption has been eliminated from its definition [4]. This point it is very important because the alcoholic parameter is measured through questionnaires and, often, is reported by individual with MASLD in a highly variable manner, from zero or near-zero to sporadic consumption during “social drinking” [4].
Worldwide, MASLD has affected 30% of the population [7]. In the next few years, its prevalence and the associated healthcare costs are expected to increase due to the spread of obesity and diabetes [3].
The multifactorial pathogenesis in which metabolic, genetic, and environmental risk factors exist has been widely explored, especially on the steatogenic and fibrogenic sides. Liver inflammation involved in the onset and progression of MASLD has aroused great interest. Evidence supports the key role of the immune response in leading to fibrogenesis promoting MASH and HCC development [8,9,10].
In MASLD, obesity and diabetes act as triggers of the inflammatory response: IR and lipotoxicity activate the hepatic immune system, and the persistent pro-inflammatory state contributes to fibrogenesis and carcinogenesis [11].
The liver hosts several resident immune system cells, such as Kupffer cells (KCs), natural killer cells (NK cells), natural killer T cells (NKT cells), mucosal-associated invariant T cells, γδ T cells, conventional CD8+ T and CD4+ T cell subsets (Th1, Th2, Th17, and T Reg cells) [12].
Different regulatory pathways counterbalance the activation of the immune system. Between them, the programmed cell death protein 1 (PD-1) represents an immune checkpoint surface receptor, acting as a key regulator of T cells and macrophage responses during chronic inflammation and cancer [13].
PD-1 activation keeps the peripheral immune tolerant and promotes the immune escape from cancer. The aim of this review is to illustrate the main pathogenetic mechanisms by which the immune system promotes the progression of MASLD and the transition to HCC and to discuss the possible therapeutic applications of PD-1/PD-L1 target therapy.

2. Innate and Adaptative Immunity in the MASLD

The immune system is essential in maintaining liver homeostasis and responding to inflammatory stimuli. The hepatic immunological environment involves a complex multistage network and interactions established between liver-resident cells and peripheral leukocytes. In this context, innate and adaptative immune mechanisms support hepatic inflammation in MASLD development and progression. However, it is largely unexplored how immune responses link with MASLD pathogenesis, but, to date, lobular inflammation is considered the driving force for MASH, fibrosis, cirrhosis, and HCC development.

2.1. The Major Immune Cell Types in the Phisiological Status and in the Early Stage of MASLD

Hepatic innate immune cells consist of a large population of macrophages, such as resident KC and lymphocytes such as NK and NKT cells, while adaptive immunity includes humoral immunity mediated by B cells and cellular immunity mediated by T cells [11,14].
Both B and T cells maximize immune surveillance, participating in maintaining tissue homeostasis and inducing systemic tolerance. The hepatic immunological tolerance involves complex and multistage interactions between liver-resident cells and peripheral leukocytes (Figure 1).
In a healthy liver, in response to acute tissue injury, the initial phase of hyper-inflammation is followed by its resolution and by anti-inflammatory phases.
Under physiological conditions, immunological homeostasis is maintained by the hepatic cytokine milieu, the balance between basal pro-inflammatory cytokines and anti-inflammatory cytokines [15], and parenchymal and nonparenchymal cells, like hepatocytes, hepatic stellate cells (HSCs), and liver sinusoidal endothelial cells (LSECs) (Figure 1) [16]. In this context, monocyte-derived cells can differentiate into liver DCs or monocyte-derived macrophages (MoMFs). The latter do not contribute to the pool of local resident macrophages but, together with KCs, which represent the “self-renewing pool”, play a role in MASLD progression, modulating hepatocyte fate (Figure 1) [17].
In the early stage of MASLD, the excessive hepatic accumulation of lipid, triggers the release of Damage-Associated Molecular Patterns (DAMPs), which are recognized by Toll-Like Receptors (TLRs), mainly TLR 1–5, expressed on the hepatic cells. It has been reported that, in MASLD patients, the expression of these TLRs is significantly upregulated [18]. These receptors are the primary sensors detecting DAMPs and triggering the activation of pro-inflammatory pathways, such as NF-κB and mitogen-activated protein kinase (MAPK) [19].
However, the concomitant state of lipid overload into hepatocytes, IR, and lipotoxicity establishes chronic inflammation, characterized by constant activation of immune responses, without adequate resolution of inflammation [20].
KCs, the hepatic resident macrophages, represent the first responders to the inflammatory stimuli in MASLD [21]. Specifically, the expression of TLR4 on KCs has been shown to be increased in MASLD patients with a diagnosis of fibrosis [22].
KCs are also involved in MASLD progression, through the release of pro-inflammatory cytokines, triggering a crosstalk with hepatic sinusoid endothelial cells, HSCs, neutrophils, monocytes, T cells, and dendritic cells (DCs) [23]. It has been reported that KCs upregulate ICAM-1 to attract more neutrophils into the liver; indeed, the serum ICAM-1 level was significantly higher in the patients with MASH than in the patients with simple steatosis and in the normal subjects [24]. In addition, KCs acting as an APC, play a tolerogenic role by expressing Programmed cell Death 1-Ligand 1 (PD-L1), an inhibitory molecule that engage PD-1, inhibits T cell-mediated immunity, and induces the activity of T Reg cells; the last cells protect against deregulated immunity responses [25]. On the other hand, KCs, during extensive injuries, increase the MoMFs influx, which replaces the native macrophages and activates naïve CD4+ T cells [26].
NK cells, one of the major populations of hepatic lymphocytes, representing about 30–50% of them, play a crucial role in the development of hepatic inflammation, antiviral responses, and tumor surveillance, through their cytolytic activity and the release of immunomodulatory cytokines. Furthermore, NK cells can kill activated HSCs, in an NKG2D-retinoic acid-dependent manner, thereby improving liver fibrosis [27].
It has been reported that, in obese subjects, there is a significant increase in NK cells with an activated phenotype and impaired ability to release granules and cytokines, which may contribute to the liver fibrosis development in MASH patients [28].
Natural killer T cells (NKT), the innate-like lymphocytes, are characterized by the expression of both T and NK cell surface markers [29]. NKTs also have a role in hepatic lipid metabolism, activating, via the expression of CD1d, a lipid antigen receptor [30]. It has been observed that CD1d deficiency impairs metabolic parameters and induces hepatic steatosis [31]. Furthermore, during acute liver inflammation, activated by CD1d, NKTs play an immunoregulatory role through the secretion of IL-4, TNF-α, and INF-γ [32]. During the early stage of MASLD, it has been shown that the number of NKTs is reduced; however, in patients with moderate-to-severe steatosis, it an increase in NKTs has been shown, with a pro-inflammatory phenotype able to promote liver fibrosis via osteopontin production [33]. So the liver attempts to trigger inflammatory response and repair mechanisms, but at the same time, the inflammation drives disease progression [34,35].

2.2. Role of the Major Immune Cell Types in the Late Stage of MASLD

Growing evidence indicates that APCs, supported by neutrophils, are a critical component of innate and adaptative immune mechanisms, which are able to activate naïve T cells. This role is not only played by professional APCs, such as LSEC, KCs, and HSCs, but also by hepatocytes [36,37].
The neutrophils contribute to tissue damage, through the production of toxic molecules, such as proteases, ROS, cytokines, and Neutrophil Extracellular Traps (NETs) [38,39,40]. The last are active participants in the inflammatory environment responsible for MASLD progression to MASH; their generation is independent of neutrophil density but is correlated with their activated state [41].
Subsequently to neutrophils, macrophages and monocytes are rapidly recruited into the liver [42].
Initially, macrophages mediate NET efferocytosis, restoring tissue homeostasis after tissue injury; secondarily, they generate an inflammatory milieu that leads to excessive neutrophil activation and NETosis induction [43].
NETs are areas dense in IL-1β and IL-17A cytokines that contribute to inflammation and correlate with MASLD progression. In this context, IL-17A can activate HSC and promote fibrosis [44]. Furthermore, it has been reported that NETs are involved in MASH-associated HCC development through the generation of immunosuppressive microenvironments. It has been demonstrated that NETs promote T Reg differentiation via the metabolic reprogramming of naïve CD4+ T-cells, promoting tumor development by suppressing cancer immunosurveillance [45].
Clinical evidence has reported the effect of NETs on the progression of MASH to HCC. Specifically, elevated levels of NET markers, such as circulating Myeloperoxidase (MPO)-DNA complexes, have been found in the serum of patients with a histological diagnosis of MASH compared to patients with normal liver histology.
During MASLD progression, in addition to neutrophils, the macrophagic population is fully activated, together with T Reg and cytotoxic CD8+ lymphocytes (Figure 2) [46]. It has been reported that the inhibition of hepatic macrophages can attenuate inflammation and the severity of liver damage, underling the crucial role of these cells in liver disease progression [47].
In the injured liver, it has been reported that monocytes can differentiate into inflammatory, angiogenic, and fibrogenic macrophages and can activate HSC or other precursor cells mediating the progression of MASH to fibrosis [48].
In this context, continuous regenerative response leads to the chronic activation of HSCs, collagen deposition, fibrosis, and subsequently cirrhosis with a switch from the fibrotic phenotype to the pro-carcinogenic one (Figure 2) [49].

2.3. Role of the Adaptative Immune Response in MASH Progression

Recent evidence suggests a main role of adaptive immunity in MASH progression since lymphocytic infiltration has been frequently observed in liver biopsies of MASH patients, often as focal aggregates of T cells, similar to ectopic lymphoid structures [9,50,51].
It has been reported that MASH is characterized by the hepatic lobular infiltrate cellular, represented mainly by of B cells, CD4+ cells, and CD8+ cells [9].

2.3.1. The Humoral Immunity: B Lymphocytes

B cells comprise two cellular subpopulations: B1, which acts through the secretion of natural antibodies, and B2, which acts through the release of high-affinity antibodies [52].
The role of hepatic B cells is not completely understood, since they comprise only 5% of human intrahepatic lymphocytes in the healthy liver. Bruzzi S and colleagues have reported that B cells are detectable in a 63% liver biopsy from patients with MASH within inflammatory infiltrates, and it has been reported that B-cell depletion ameliorated the hepatic inflammatory response status [9,53,54].
After their stimulation, B2 cells are activated and express high levels of B Cell Activating Factor (BAFF or CD257); its receptor, BAFF-R, a tumor necrosis factor family member, is expressed on the surface of monocytes, macrophages, dendritic cells, neutrophils, and activated T cells; it is responsible for their activation [55]. In mouse models of MASLD, BAFF neutralization has been able to reduce steatohepatitis [53].
It has been reported that MASH patients have higher circulating levels of BAFF compared to those with simple steatosis. It has been suggested that serum BAFF levels could be useful for distinguishing MASH from simple steatosis. Indeed, it has been reported that, in MASH patients, an increase of 1.5 times the BAFF levels is associated with the presence of hepatocyte ballooning and advanced fibrosis [56].

2.3.2. The Cellular Immunity: T Lymphocytes

The T lymphocytes effectors of phagocytosis and of tumor killing [57] comprise three subgroups: CD4+ T cells (which include helper T (Th)1, Th2, Th17, follicular helper T (Tfh) cells, and T Reg cells), CD8+ T cells, and γδ-T cells (Figure 3) [58,59].
CD4+ T lymphocytes are preponderant effectors of adaptative response; they, in the presence of persistent pro-inflammatory stimuli, acquire a range of polarized phenotypes, which are mostly classified into Th1, Th2, Th17, and T Reg cells (Figure 3) [60,61,62].
Regarding the role of CD4+ T cells, it has been reported that Th1 and Th17 lymphocytes are prevalent in hepatic tissues and the peripherical blood of patients with MASH [63]. In MASH, Th1 cells are predominant and are induced by IFN-γ. Indeed, in IFN-γ- deficient mouse models of NASH, the inhibition of inflammatory response and the consequent protective effect against liver injury and hepatic fibrosis development have been reported [64].
TH17 cells have both homeostatic and pathogenic roles in producing cytokines such as IL-17F, IL-22, and IL-17A [65]. The latter plays an important role in MASLD progression; it has been reported that treatment with anti-IL-17 monoclonal antibody of high-fatty-diet (HFD) mice improved liver function with the attenuation of lipid accumulation. Moreover, the treatment was able to inhibit KC activation, inducing a decrement of pro-inflammatory cytokine levels [66].
An increase in the levels of Th17 has also been reported in MASH patients compared to those with MASLD [67]. Specifically, in MASH, the injured hepatocytes appear to be responsive to IL-17A signaling, upregulating the expression of its receptor IL-17RA, compared to healthy hepatocytes, where its expression is lower [68]. Several studies have reported that IL-17A can exacerbate steatosis and fibrosis by promoting hepatic DNA injury [68]. The prevalent role of Th7 signaling in MASLD progression has been confirmed in a model of HFD mice, where the administration of monoclonal antibody targeting IL-17 induced a marked decrease in hepatic lipid accumulation and attenuated liver fibrosis (Figure 2) [68].
CD8+ T cells mediate metabolic dysregulation and amplify the IR status [69]; their activation is supported by type I IFN [70].
In both humans and mouse models, the hepatic accumulation of CD8+ T lymphocytes during MASLD progression. In particular, it has been demonstrated that, in mice with MASH, CD8+ T cells induced liver fibrosis via the activation of HSCs and through the production of IL-10 and TNFα [71]. Furthermore, in response to metabolic stimuli, the CXCR6+ CD8+ T lymphocytes act as auto-aggressive cells in a MHC-class-I-independent manner, resulting in the upregulation of Fas ligand expression and subsequent hepatocyte apoptosis [10]. The pharmacologic or genetic ablation of CD8+ T cells ameliorates steatosis, IR, and inflammation (Figure 3) [72,73]. In mouse models, it has been shown that, in an inflammatory milieu, the losing of liver tolerance is mediated by KC activation and T Reg cell (T Reg cell) reduction (Figure 3) [25].
T Reg cells play an immunosuppressive role, contributing to liver tolerance in healthy liver [11]. In contrast to this, in MASLD it has been shown that T Reg cells are more susceptible to apoptosis in response to oxidative stress. Consequently, MASH patients showed a reduction in levels T Reg on the peripherical blood and in the liver, together with an imbalance between the intrahepatic levels of Th17 and T Reg cells. However, it is not clear if it is a consequence of local immune dysregulation or is mediated by extrahepatic factors (Figure 3) [67].
Immunological tolerance mechanisms include the expansion of CD4+ T Reg cells, as well as the apoptosis inhibition of autoreactive cells, a mechanism commonly described for CD8+ T cells.
The tolerance induction is related to the expression of the inhibitory molecule PD-L1, expressed on the surface of lymphocytes when the liver is healthy, whereas if it is injured, PD-L1 expression is suppressed, and the immunity system response acquires a phenotype influenced by Th1 cells. In this context, adaptive immunity mediates chronic and aggressive inflammation, inducing the increase inCD8+ T cytotoxic cell activation and the expression of the PD-1 receptor. Variations in the expression of inhibitory T cell receptors (PD-1 and 2B4) on CD8+ T lymphocytes have been linked to the immune tolerance necessary for the MASH progression [74].

3. Impact of PD-1/PD-L1 in Chronic Metabolic Diseases

3.1. The PD-1/PD-L1 Immune Pathway and the Role of CTLA-4/B7 and TIGIT/CD112 Co-Inhibitor Receptors

3.1.1. PD-1/PD-L1 Immune Pathway

Programmed cell death protein 1 (PD-1), also called PDCD1 or CD279, is a member of the B7/CD28 receptor superfamily and is a type I transmembrane protein, with a complex organization into domains and into motifs, like ITIM and ITSM, required for its immunosuppressive activity [75,76] (Figure 4).
PD-1 is activated by two ligands, which are type I transmembrane proteins and belong to the B7/CD28 family: PD-L1, and PD-L2 [75,76,77]. They differ in their affinities for PD-1: PD-L2 has a higher affinity, though PD-L1 has a wider range of expression than PD-L2 [77]. PD-L1 and PD-L2 expressions are upregulated on T and B cells by the inflammatory cytokines type 1 and type 2 interferons, TNF-α, IL-2, IL-7, IL-21, and IL-15 [78,79]. In addition, cancer cells are expressed on both PD-L1 and PD-L2, but PD-L1 is the main player in tumor immune escape.
PD-1, a coinhibitory receptor, limits the over-activation of immune responses and promotes self-tolerance through the modulation of activated T cells, the induction of antigen-specific T cell apoptosis, and the inhibition of regulatory T cell apoptosis [78]. On the other hand, the PD-1 pathway limits the immunopathological responses; indeed, whether the response of CD8+ T cells is not adequately controlled, it can result in the overproduction of pro-inflammatory cytokines, driving aberrant cell killing, tissue damage, and the development of severe immunopathology.
In recent years, PD-1 has received much attention for its roles in inducing an exhausted T cell phenotype and tumor immunosuppression. Indeed, a correlation between PD-1 signaling and metabolic activity has been reported in T cells [80]. Specifically, during T cell activation, PD-1 signaling can modulate metabolic reprogramming, inducing the switch from oxidative phosphorylation to aerobic glycolysis, becoming effector cells [81]. In addition, in CD4+ T cells, the PD-1 pathway promotes lipolysis and fatty acid oxidation [82]. In the tumor microenvironment, this metabolic competition drives tumor progression by inducing the hyper-responsivity of T cells through glucose deprivation [83].
Several studies have reported PD-1 function in different cell types, but the knowledge of its signaling and functions on activated T cells is not completely understood. PD-1 mediates inhibitory signals when engaged with the T cell receptor (TCR) or the B cell receptor (BCR) (Figure 4) [84]. It is known that in T lymphocytes, PD-L1/PD-1 signaling antagonizes CD80-CD28 costimulation, inducing the inhibition of T-cell proliferation, cytokine production, and cytolytic function and impairing T-cell survival (Figure 4) [85].
PD-1/PD-L1 pathway may inhibit T-cell function and survival by directly blocking TCR signals or, alternately, can exert an indirect inhibitory effect on CD28-costimulation by regulating the expression of CK2 and cyclin-dependent kinases (CDKs) (Figure 4) [86].

3.1.2. CTLA-4/B7: A Co-Inhibitor Receptor of Immune Response

The immune pathways are regulated not only by the PD-1/PD-L1 axis but by other ones, such as CTLA-4/B7 and TIGIT/CD112, which can contribute to the pathogenesis of MASLD and to the transitioning of HCC. CTLA-4/B7 (Cytotoxic T Lymphocyte Antigen 4) is an inhibitory immune checkpoint acting together with PD-1/PD-L1 in the regulation of physiological immune homeostasis. It downregulates inflammatory responses and can facilitate the immune evasion of cancer cells in the early stages of the disease [87]. CTLA-4 is expressed on CD4+, CD8+ T, and T Reg cells and negatively regulates their function, acting during their phases of activation. Indeed, differently from PD-1, which acts during the effector phase, impairing the T cells, proliferation and differentiation with the establishment of an exhausted phenotype, CTLA-4 is expressed on CD8+ and CD4+T cells after their stimulation, preventing the proliferation signal; conversely, it is expressed on T Reg cells in a constitutive manner [78,88,89]. CTLA-4 acts to recognizing ligands B7-1 and B7-2 (or CD80 and CD86), expressed principally on APC, and, respectively, in a manner constitutive of B7-1, whereas B7-2 acts later [90]. B7-1 and B7-2, expressed on APC, interact with the CD28 of T cells and induce the secretion of inflammatory cytokines, initiating inflammatory responses though the activation of CD8+, CD4+, and T Reg cells [91,92]. T Reg cells are considered canonical anti-inflammatory cells, and CD28 could, in this manner, indirectly contrast MASLD progression, activating hepatic T Reg accumulation.
On the other hand, when B7 interacts with CTLA-4, it induces the inhibition of naïve T lymphocytes and T Reg suppressive functions, resulting in the downregulation of the T cell response [93]. It has been reported that B7 costimulation (1 and 2) in mice fed HFD reduced the T Reg cell number, promoting MASLD progression [94].
In addition, it has been reported that, under conditions of low inflammation, such as in the early stages of tumorigenesis, APC cells express low levels of B7 molecules and CTLA-4 activity is lower, so it cannot inhibit the T cell response and allows them to participate in the antitumoral response [95]. In contrast, in conditions of chronic inflammation, during tumorigenesis progression, the activation of innate and adaptive immune cells can increase the expression of costimulatory molecules, such as B7, which, when interacting with CTLA-4, downregulates T cell activation [87]. In this manner, CTLA-4/B7 prevents the elimination of immune-mediated tumoral cells, favoring the development of immunological tolerance against them [96].
To date, immune checkpoint inhibitors (ICI) have been designed to be able to act against CTLA-4/B7 and PD-1/PD-L1pathways, and the efficacy of combined immunotherapy with MoAb in cancer treatment has recently been applied, showing higher clinical efficacy compared to either agent individually [97].
Since CTLA-4 plays a critical role in the downregulation of T cell responses at the beginning of the immune response, its inhibition, mediated by its blockade, could be a strategy for the enhancement of T cell responses in immunotherapy, especially in the early stage of tumorigenesis. Interest in these important immune checkpoint pathways arises from their role in modulating physiological immune homeostasis, downregulating the inflammatory response, and facilitating the immune evasion of tumor cells [98].

3.1.3. TIGIT/CD112: A Co-Inhibitor Receptor of Immune Response

Another newly identified coinhibitory receptor is the T-cell Ig and ITIM domain (TIGIT) expressed by activated CD8+ T and CD4+ T cells, NK cells, T Regs cells, and the follicular T helper [99]. The ligands of TIGIT are CD112 and CD155, which are expressed by tumor cells and APC. CD155, also called Poliovirus Receptor (PVR), shows a high affinity for TIGIT, whereas CD112 is a low-affinity ligand for TIGIT biding. Interestingly, CD155 and CD112 are expressed by a variety of solid tumors, including liver cancer [100]. Recently, a novel coinhibitory receptor for T cells has been identified, named CD112R [101].
Similar to the CD28/CTLA4 pathway, TIGIT/CD112 constitutes an emerging signaling pathway relating to the regulation of T cell immunity; indeed, a TIGIT blockade increases CD8+ T cell expansion against tumor antigens [102]. In cancer, TIGIT is expressed, together with PD-1, on tumor antigen-specific CD8+ T cells and on exhausted CD8+ T cell subsets and is associated with constant inflammatory responses [103].
TIGIT acts to inhibit innate and adaptative immunity, indirectly blocking T cell function by binding to CD155 on DC and directly reducing T cell proliferation [104]. Indeed, TIGIT binds CD155 expressed on CD8+ T cells and NK cells, with higher affinity limiting their activation. In this manner, TIGIT impairs antitumoral immunity and sustains cancer development [105]. TIGIT blockade has been shown to increase antitumor NK cell activity and CD8+ T cell cytokine production/cytotoxicity; the antitumor T cell effect was NK-cell-dependent. In this context, the PD-1 and TIGIT blockades synergize to augment the proliferation and function of CD8+ T cells with antitumoral effects [106].
Considering the prevalent role of inhibitor receptors in regulating innate and adaptative immunity in chronic disease, such as MASLD, characterized by the hepatic progressive accumulation of exhausted CD8+ T cells, and in cancer, where the dysfunctional/exhausted T cells are prevalent, the upregulation of CTLA-4/b7 and TIGIT/CD112 checkpoint inhibitory receptors limits T cell survival and function. In this scenario, their interaction with the PD-1/PD-L1 pathway could amplify the mechanisms of immune escape, favoring MASLD progression or HCC development.

3.2. PD-1/PD-L1 Axis in Obesity

Obesity is a major risk factor for MASLD development and is responsible for low-grade inflammation in white adipose tissues (WATs) [107].
WAT plays an important role in energy storage and produces several biologically active molecules, such as adipokines, that act as regulators of systemic metabolism and immune modulators.
WAT is colonized by a wide range of leukocyte populations: CD4+, CD8+, Treg, B cells, iNKT, mast cells, eosinophils, innate lymphoid type 2 cells (ILC2s), macrophages, and DCs [107,108]. The resident adipose leukocyte disruption imbalances homeostasis, contributing to the development of obesity-associated inflammation and metabolic disorders.
In the initial stages of the inflammatory state, excessive nutrient intake causes the pathologic expansion of adipose tissue, which results in cell hypertrophy, the failure of lipid storage, hyperinsulinemia, dyslipidemia, and adipocyte hypoxia [109]. This persistent condition of oxidative stress and inflammatory stimuli leads to adipocyte apoptosis and the release of pro-inflammatory and chemotactic mediators and consequently leukocyte infiltration. Adipose inflammation leads to CD8+ T cell activation, which propagates inflammatory signals and shifts the M2 phenotype of macrophages into M1 [108].
The M1 cells form crown-like structures (CLSs) around the apoptotic adipocytes, resulting in WAT inflammation [110].
In inflamed adipose tissue, persistent T-cell antigen receptor stimulation induces CD8+ T cells to acquire an exhausted phenotype and consequently the loss of proliferative activity and cytotoxic functions. Eljaafari et al. have demonstrated that in mice and obese humans, the over-expression of PD-1 is responsible for exhausted T cells [111]. Shirakawa and colleagues demonstrated that in obese WATs, there is a unique subpopulation of CD153+, PD-1+, CD44hi, CD4+, and T lymphocytes that is linked with WAT inflammation and systemic insulin resistance [112]. In a murine model of obesity, PD-1 is an intrinsic negative regulator of ILC2 function in the presence of PD-L1+ macrophages. TNF seems to play a central role in the disruption of homeostasis by the PD-1/PD-L1 pathway, triggering IL-33, and M1 activation [113]. In a knockout mouse model, PD-L1 expression limits adipose tissue inflammation and obesity with the increase in Th1 cells and a reduction in ILC2 and Treg cells [114].
Obesity in NASH patients represent an important risk for HCC development. Recent studies have shown that cytokines produced by the inflammasome may contribute to the inflammatory microenvironment in obesity, which promotes NASH progression and HCC [115].
Data indicate that the NLRP3 inflammasome is stimulated by lipotoxicity and higher levels of IL-1β. In high-fat-fed (HFF) portal veins of mice, IL-1β is elevated [116].

3.3. PD-1/PD-L1 Axis in Type 2 Diabetes

Type 2 diabetes mellitus (T2D) is a complex chronic disorder that results from the dysregulation of lipids, protein, and carbohydrate metabolism, resulting in impaired insulin secretion and insulin resistance. A PD-1/PD-L1 signaling pathway is associated with T2D, which has proven to be a vital target for its therapy [117].
In patients with T2D, PD-1 is less expressed. It has been reported that the serum of these patients contains soluble costimulatory and cytokines, which influence PD-1 expression and function [118]. The induction of B-cell activation and cytokine production by follicular helper T cells (Tfh) is linked to the persistent low-grade inflammatory state that is characteristic of T2D. Indeed, in the evaluation of circulating CD4+CXCR5+ T cells (CTfh) in the peripheral blood of T2DM patients, a significant increase in the number of CTfh has been observed in the peripheral CD4+ T cells compared to controls. Additionally, an imbalance in the CTfh subtypes and an increase in the Th17 subtype have been reported in T2DM patients. Further analyses have revealed that CTfh levels were significantly high in patients with a body mass index (BMI) over 24.0. Interestingly, patients with abdominal obesity showed a further increase in CTfh levels compared to those without abdominal obesity. This finding suggests a potential association between CTfh and T2DM-related obesity. Another study has reported that PD-1 was more expressed in CD4+ T cells from healthy controls but not in T2D patients. A low expression level of PD-1 can result in abnormal T cells and the progression of T2D [119].
Nishimura et al. have demonstrated that CD8+ T cells are in charge of macrophage activation, and their polarization from M2 to M1 mediated by PD-1 triggers the progression of T2D into obese mice [108,120].
The number of NK cells has also been found to be higher in T2D patients [121]. T2D also impairs the function of NK, which is an important target for infection and tumor protection.
In tumor immunity, PD-1 upregulation can mediate the exhaustion of activated NK cells. PD-1 is expressed on NK cells in T2D patients and is less expressed in healthy donors. It has been reported that monocytes isolated from T2D patients have a low expression of PD-1, demonstrating that T2D patients display an altered insulin sensitivity [118].

3.4. The PD-1/PD-L1 Axis in MASLD

MASLD is a multifactorial disease, and its exact pathogenesis is not completely understood. Several interlinked processes contribute to inducing liver injury, steatosis, and inflammation, which together progress to fibrosis and, sometimes, to HCC. In recent years, PD-1 and PD-L1 checkpoint inhibitors have been used as therapy for HCC; indeed, immunotherapy is thought to activate T cells and reinvigorate immune surveillance against cancer. Recently, a relationship between the efficacy of immunotherapy and HCC etiologies has been underlined and consequently the importance of prognostic factors that allow an optimal response to therapy [122].
A meta-analysis study on the effect of inhibitors of PD-L1 or PD-1 in patients with HCC revealed that the immune therapy did not improve survival in patients with non-viral HCC. Moreover, patients with NASH-driven HCC who received anti-PD-1 or anti-PD-L1 treatment showed reduced survival compared to patients with other etiologies [122].
Recent data have reported that the hepatic progressive accumulation of unconventionally exhausted resident cytotoxic T cells (CD8+ PD1+ CXCR6+), which are characterized by a tissue residency (CXCR6), effector (granzyme), and exhausted (PD-1) phenotype [10].
The mechanism driving T cell exhaustion has been attributed to continuous antigen exposure, together with the hypoxic condition in the hepatic environment, leading to mitochondrial dysfunction [123]. Although the elimination of persistent stimulus can rescue terminally exhausted T lymphocytes, progenitor exhausted CD8+ T cells, which maintain their capacity to secrete inflammatory cytokines, maintain a molecular exhaustion sign (Figure 5).
Ongoing T lymphocyte activation and the concomitant persistent expression of PD-1, associated with exhausted phenotype, are related to the loss of T cell activity and tumor escape from immune responses.
Moreover, it has been observed in preclinical models and patients of MASLD or MASH treated with PD-1-direct antibodies that CD8+ PD1+ CXCR6+ lymphocytes showed impaired hepatic immune surveillance, and they also contribute to the progression of tissue damage [122] (Figure 3). Tissue damage was related to an increase in hepatic CD8+ PD1+ CXCR6+ T cells. Instead, the depletion or neutralization of CD8+ PD1+ CXCR6+ lymphocytes attenuates the progression of hepatic cancer.
Interestingly, in the setting of excessive fat accumulation and lipotoxicity, the PD1 checkpoint inhibitors treatment leads to an expansion of intra-tumoral T lymphocytes (TIL), exacerbating liver damage and promoting tumor progression. Fatty liver disease likely seems to inhibit immune responses against HCC.
In the setting of metabolic fatty liver disease, the crucial role of immune system dysregulation and, in particular, of cytotoxic lymphocytes has been highlighted; indeed, the hepatic accumulation of CD8 T cells with phenotypes that combined tissue residency (CXCR6) with effector (granzyme) and exhaustion (PD1) characteristics has been detected [10,122].
Several studies have reported the association of polymorphisms in PD-1 and PD-L1 genes with a higher risk of cancer development [124]. The correlation between the single-nucleotide polymorphism (SNP) rs7421861 of PDCD1 with higher PD-1 expression on monocytes and T lymphocytes has been demonstrated, demonstrating a potential role for PD1 in the susceptibility of MASLD to HCC [125,126].

Pro- and Anti-Inflammatory Effects of PD-1/PD-L1 Axis in MASLD Progression

The lipotoxicity-mediated damage to hepatocytes involves the activation of innate and adaptive immune responses. During the initial phase of inflammation, CD4+ effector T cells are activated first, and only later do NKT cells and CD8+ T cytotoxic lymphocytes contribute to tissue injury [34]. As a consequence of T cell activation, lymphocytes and liver cells overexpress PD-1 and PD-L1 as a self-defense mechanism to limit the activity of T cells [127].
The role of PD-1 in inflammation in MASLD and MASH is still not properly understood. However, even with the lack of published studies relating to its involvement, links from other inflammatory-related diseases and known inflammatory factors can be translated to MASLD and to MASH.
In the initial stage, chemokines and growth factors stimulate and activate T cells. However, chronic antigen exposure leads T cells to acquire an exhaustion phenotype with consequent PD-1 upregulation [85]. In this context, both CD4+ and CD8+ T cells can be differentiated in cellular subsets, contributing to the establishment of a pro- or anti-inflammatory status. Among these, Th1 and Th17 CD4+ T cells mediate a pro-inflammatory role, whereas Th22 and Treg CD4+ T cells play an anti-inflammatory role [22].
In the different phases of the inflammatory process, the upregulation of PD-1 in the Th1 or Th17 CD4+ T cells leads to the resolution of inflammation, whereas the overexpression of PD-1 in the Th22 or Treg CD4+ T cells amplifies the pro-inflammatory status, which, together with CD8+ T cells, is responsible for the progression from MASLD to NASH [63]. However, it has been reported that perforin-secreting CD8+ T cells protect against hepatic inflammation in mice with MASH.
On the other hand, the role of ligands of PD-1 and PD-L1 in MASLD and NASH is better understood. During the chronic lipotoxicity-mediated injury of hepatocytes, the activation of KCs is mediated by TLR receptors [128]. KCs represent the first line of defense and, when activated, they release pro-inflammatory factors, such as TNF-α, IL-1β, IL-6, IL-12, and IL-18 [21]. These cytokines and chemokines upregulate the expression of PD-L1 [97]. The overexpression of PD-L1 prolonged the inflammation within the liver, resulting in an upregulation of both PD-1 and PD-L1. The activation of PD-1/PD-L1 initially acts as a response to limit damage to the liver tissue caused by the immune system, but secondarily, their persistent activation contributes to the progression of MASLD.

4. Potential Role of PD-1/PD-L1 as Biomarkers in MASLD

No biomarkers related to the PD-1/PD-L1 axis are currently available for helping clinical practice, but the relevance of the PD-1/PD-L1 axis in the pathogenesis of MASLD suggests the intriguing possibility of using these molecules as biomarkers for the early identification of disease and for assessing the risk of progression, especially in the onset of HCC development in MASLD.
However, two major challenges should be addressed. First is the feasibility and reliability of measuring PD-1/PD-L1 expression in clinical settings. Second is how utilizing these biomarkers might impact early intervention strategies or patient management. A recent study [126] showed a lack of correlation between a PD1 expression and PD1 rs13023138 G variant. This may significantly complicate the search for reliable biomarkers. This may be particularly true when the involved variant is intronic, as not all genetic polymorphisms or intronic variants directly translate into relevant changes in protein expression or biological function. This scenario underscores the need for a more in-depth and detailed analysis of the different genetic variants involved in the PD-1/PD-L1 axis and their effect on protein expression and biological function to identify more reliable and clinically useful biomarkers for MASLD.

5. The Rationale of Immunotherapy in MASLD

The complex and central role of the immune system in the pathogenesis and progression of MASLD sheds light on the potential use of immunomodulation as a target therapy.
Although many drugs against MASLD are under investigation, no pharmacological therapy has been approved so far, and lifestyle changes play a dominant role in clinical practice [129].
The full spectrum of molecules acting as chemokine and nuclear receptors, tyrosine-kinases, and molecules expressed on the surface of immune-system cells are currently in use as a target therapy in MASLD. The chemokine (C-C motif) ligand (CCL) 2/C-C chemokine receptor 2 (CCR2) pathway recruits Ly-6Chi monocytes, and it encourages the phenotypic switch from Ly-6Chi macrophages to Ly-6C low macrophages that are the main source of MMPs promoting fibrosis resolution [130,131]. The chemokine (C-C motif) ligand (CCL) 2/C-C chemokine receptor 5 (CCR5) acts as a regulator of migration, activation, and proliferation of collagen-producing activated HSCs/myofibroblasts [132]. Cenicriviroc is an antagonist of CCR2/CCR5 chemokine receptors on pro-inflammatory monocytes. It showed an improvement in fibrosis in patients with histological MASLD versus a randomized vs. placebo, controlled, phase IIb clinical trial (CENTAUR trial). However, these results were not further confirmed in the randomized vs. placebo, controlled, phase III trial (AURORA trial), and the development of the drug was discontinued [133].
Peroxisome proliferator-activated receptors (PPARs), a subfamily of the NR1C nuclear receptors, are ligand-activated transcription factors. The three isotypes, PPARα (NR1C1), PPARδ (PPARβ or NR1C2), and PPARγ (NR1C3), have different tissue-distribution patterns and functions, playing a key role in the liver as regulators of glucose and lipid metabolisms, inflammation through macrophage regulation and fibrosis, keeping hepatic stellate cells quiescent [134]. Elafibranor (GFT505), a dual agonist of PPARα and PPARδ, was shown to be promising in the randomized versus placebo, controlled, phase IIb clinical trial (GOLDEN-505 trial). The post hoc analysis showed that the administration of 120 mg of Elafibranor resolved NASH without worsening fibrosis in the intention-to-treat analysis and in patients with moderate or severe MASH. However, the predefined endpoint was not met in the intention-to-treat population. However, Elafibranor failed to achieve the primary endpoint in the randomized versus placebo, controlled, phase III trial (RESOLVE-IT trial) [135]. Lanifibranor, a pan-PPAR agonist, resulted in a decrease of at least two points in the SAF-A score without a worsening of fibrosis compared to placebo in MASLD in a randomized, controlled, phase IIb clinical trial (NATIVE trial), supporting the further assessment of Lanifibranor in phase III trials [136].
Farnesoid X receptor (FXR) is a nuclear receptor expressed in the liver, with two known variants, FXRα (NR1H4) and FXRβ (NR1H5). Upon ligand activation, FXR binds to the transcriptional responsive elements as either monomers or heterodimers with retinoid X receptor (RXR), and it seems to exert an anti-inflammatory and anti-fibrotic effect and regulate glucose and lipid metabolism [129].
Obeticholic acid (OCA) is a 6α-ethyl derivative of chenodeoxycholic acid (CDCA) and represents the first-in-class selective steroid FXR agonist. It showed an improvement of fibrosis in histological MASLD in the randomized, controlled phase IIb clinical trial (FLINT trial) [137]. The randomized, controlled, phase III clinical trial (REGENERATE trial) revealed a potential effect of OCA on improving fibrosis compared to placebo but without any effect on MASH resolution, which did not encourage the approval of the drug by the FDA [138].
Tropifexor is an FXR agonist that was used alone and in combination with Cenicriviroc in histological MASH in a randomized versus placebo, controlled, phase IIb clinical trial (TANDEM trial). The safety profile of the combination was similar to respective monotherapies, and no substantial incremental efficacy was observed with the combination on ALT, body weight, or in histological end points compared with the monotherapy [139].
Monoclonal antibodies targeting T cell receptor-associated molecule CD3 represent a new horizon in MASLD treatment. However, the use of the murine monoclonal antibody (OKT3) is limited due to the toxicity shown in a phase IIa clinical trial, and a new fully human anti-CD3 antibody, Foralumab, already tested for Chron’s Disease and in renal allograft rejects, is going to be tested in a setting of MASLD and type 2 diabetes patients [140].
The spectrum of the potential treatment of MASLD through the immunomodulation is huge, but none of the treatments have been fully convincing so far. Further efforts are required to understand the pathogenesis and to control the progression of MASLD. Moreover, we cannot rule out the possibility of combination immunotherapy that might be the new frontier in MASLD treatment in the future.

6. Limitations

The clinical trials currently present in the literature demonstrate how the PD-1/PD-L1 axis could be used for the treatment of MASLD. However, the pathogenesis of MASLD is complex, involving multiple risk factors such as metabolic factors such as obesity and diabetes in a bidirectional manner, leading to lipotoxic damage, inflammatory response, and progressive deposition of fibrotic tissue. This could be a major limitation of immunotherapy, as it targets only a small fraction of the factors involved in the onset and progression of the disease, potentially accounting for the inadequate action of drugs tested thus far. Likely, a combination therapy might hold the key to optimizing the efficacy of drugs acting on the PD-1/PD-L1 axis. Although patients in such trials were correctly randomized, biases in patient’s enrollment, centralized biopsies reading, inadequate sample size, and short follow-up might influence the outcomes.

7. Conclusions

The dysregulation of the immune system plays a crucial role in the pathogenesis of MASLD and its progression to HCC.
In obesity and T2D, the PD-1/PD-L1 pathway mediates the dysfunction of hypertrophic adipocytes, M2-to-M1 polarization, and IL-33 production, maintaining inflammation of WAT, and inducing systemic insulin resistance. In MASLD progression, mitochondrial dysfunction and lipotoxicity upregulate PD-1, which results in the exhaustion of resident cytotoxic cells. In the tumoral context, PD-1-PD-L1 signaling mediates the exhaustion of activated NK cells and T lymphocytes. The continuous activation of T lymphocytes promotes tumor cells to escape from the immune response.
To date, it has been reported that immunotherapy improves survival only in patients with viral HCC compared to non-viral ones, whereas in MASLD-related HCC patients, the blockage of PD-1/PD-L1 signaling is associated with the decreased survival of patients.
PD-1/PD-L1 inhibitors play a critical role in cancer. Several monoclonal antibodies act as inhibitors to disrupt the interaction between PD-1 and PD-L1. In 2018, the FDA approved therapy, which only treats patients with metastasis or those who are not candidates for surgery or radiotherapy. Hence, studying the use of immunotherapy to block the PD-1/PD-L1 axis represents a complex goal for future study.

8. Future Directions

Although drugs targeting the PD-1/PD-L1 axis show significant promise in both MASLD and HCC, discussing future perspectives remains challenging. The pathogenesis of MASLD is complex and likely not entirely elucidated, and the treatment is equally complex and hardly standardizable. Large-scale population studies with follow-ups of at least three years are undeniably necessary to better understand the efficacy of these treatments.
As previously indicated, identifying optimal drug combinations targeting PD-1/PD-L1 and drugs with other mechanisms of action is crucial to develop therapies addressing multiple targets, potentially representing the future of MASLD.

Funding

The research that led to these results received funding from MIUR within the PNRR M4C2I1.3 Heal Italia project PE00000019 CUP B73C22001250006 to Salvatore Petta. The project is supported by the Italian project PNRR-MAD-2022-12375656. The project is supported by PRIN 2022 2022L273C9. The project is supported by RF-2021-12372399.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

APCAntigen-presenting cell
CCRChemokine (C-C motif) ligand (CCL) 2/C-C chemokine receptor
CDCluster of differentiation
CTLA-4Cytotoxic T Lymphocyte Antigen 4
DCDendritic cell
HCCHepatocellular carcinoma
HSCHepatic stellate cells
ICIImmune Checkpoint Inhibitors
IgImmunoglobulin
ILC2Innate lymphoid type 2 cell
IFNInterferon
ILInterleukin
IRInsulin resistance
KCKupffer cell
LSECLiver sinusoidal endothelial cell
MoMfMonocyte-derived macrophage
MASLDMetabolic-associated fatty liver disease
MASHMetabolic dysfunction-Associated Steatohepatitis
NASHNonalcoholic steatohepatitis
NETsNeutrophil Extracellular Traps
NKNatural killer
NKTNatural killer T
OCAObeticholic acid
PPARPeroxisome proliferator-activated receptor
PD-1Programmed cell death protein 1
PD-L1Programmed cell death ligand 1
T2DType 2 diabetes mellitus
TGFTransforming growth factor
THT helper
TIGITT-cell Ig and ITIM domain
TLRToll-like receptor
TNFTumor necrosis factor
T RegT regulatory
WATWhite adipose tissue

References

  1. Vitale, A.; Svegliati-Baroni, G.; Ortolani, A.; Cucco, M.; Dalla Riva, G.V.; Giannini, E.G.; Piscaglia, F.; Rapaccini, G.; Di Marco, M.; Caturelli, E.; et al. Epidemiological Trends and Trajectories of MAFLD-Associated Hepatocellular Carcinoma 2002–2033: The ITA.LI.CA Database. Gut 2023, 72, 141–152. [Google Scholar] [CrossRef]
  2. Pennisi, G.; Enea, M.; Romero-Gomez, M.; Viganò, M.; Bugianesi, E.; Wong, V.W.-S.; Fracanzani, A.L.; Sebastiani, G.; Boursier, J.; Berzigotti, A.; et al. Liver-related and Extrahepatic Events in Patients with Non-alcoholic Fatty Liver Disease: A Retrospective Competing Risks Analysis. Aliment. Pharmacol. Ther. 2022, 55, 604–615. [Google Scholar] [CrossRef]
  3. Pennisi, G.; Pipitone, R.M.; Enea, M.; De Vincentis, A.; Battaglia, S.; Di Marco, V.; Di Martino, V.; Spatola, F.; Tavaglione, F.; Vespasiani-Gentilucci, U.; et al. A Genetic and Metabolic Staging System for Predicting the Outcome of Nonalcoholic Fatty Liver Disease. Hepatol. Commun. 2022, 6, 1032–1044. [Google Scholar] [CrossRef]
  4. Eslam, M.; Sanyal, A.J.; George, J.; Sanyal, A.; Neuschwander-Tetri, B.; Tiribelli, C.; Kleiner, D.E.; Brunt, E.; Bugianesi, E.; Yki-Järvinen, H.; et al. MAFLD: A Consensus-Driven Proposed Nomenclature for Metabolic Associated Fatty Liver Disease. Gastroenterology 2020, 158, 1999–2014.e1. [Google Scholar] [CrossRef]
  5. Younossi, Z.M.; Golabi, P.; Paik, J.M.; Henry, A.; Van Dongen, C.; Henry, L. The Global Epidemiology of Nonalcoholic Fatty Liver Disease (NAFLD) and Nonalcoholic Steatohepatitis (NASH): A Systematic Review. Hepatology 2023, 77, 1335–1347. [Google Scholar] [CrossRef]
  6. Wattacheril, J. Extrahepatic Manifestations of Nonalcoholic Fatty Liver Disease. Gastroenterol. Clin. N. Am. 2020, 49, 141–149. [Google Scholar] [CrossRef]
  7. Younossi, Z.M.; Koenig, A.B.; Abdelatif, D.; Fazel, Y.; Henry, L.; Wymer, M. Global Epidemiology of Nonalcoholic Fatty Liver Disease—Meta-Analytic Assessment of Prevalence, Incidence, and Outcomes. Hepatology 2016, 64, 73–84. [Google Scholar] [CrossRef]
  8. Huby, T.; Gautier, E.L. Immune Cell-Mediated Features of Non-Alcoholic Steatohepatitis. Nat. Rev. Immunol. 2022, 22, 429–443. [Google Scholar] [CrossRef]
  9. Sutti, S.; Albano, E. Adaptive Immunity: An Emerging Player in the Progression of NAFLD. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 81–92. [Google Scholar] [CrossRef]
  10. Dudek, M.; Pfister, D.; Donakonda, S.; Filpe, P.; Schneider, A.; Laschinger, M.; Hartmann, D.; Hüser, N.; Meiser, P.; Bayerl, F.; et al. Auto-Aggressive CXCR6+ CD8 T Cells Cause Liver Immune Pathology in NASH. Nature 2021, 592, 444–449. [Google Scholar] [CrossRef]
  11. Heymann, F.; Tacke, F. Immunology in the Liver—From Homeostasis to Disease. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 88–110. [Google Scholar] [CrossRef]
  12. Peiseler, M.; Tacke, F. Inflammatory Mechanisms Underlying Nonalcoholic Steatohepatitis and the Transition to Hepatocellular Carcinoma. Cancers 2021, 13, 730. [Google Scholar] [CrossRef]
  13. Sharpe, A.H.; Pauken, K.E. The Diverse Functions of the PD1 Inhibitory Pathway. Nat. Rev. Immunol. 2018, 18, 153–167. [Google Scholar] [CrossRef]
  14. Bogdanos, D.P.; Gao, B.; Gershwin, M.E. Liver Immunology. In Comprehensive Physiology; Terjung, R., Ed.; Wiley: Hoboken, NJ, USA, 2013; pp. 567–598. ISBN 978-0-470-65071-4. [Google Scholar]
  15. Robinson, M.W.; Harmon, C.; O’Farrelly, C. Liver Immunology and Its Role in Inflammation and Homeostasis. Cell Mol. Immunol. 2016, 13, 267–276. [Google Scholar] [CrossRef]
  16. Crispe, I.N. The Liver as a Lymphoid Organ. Annu. Rev. Immunol. 2009, 27, 147–163. [Google Scholar] [CrossRef]
  17. Varol, C.; Yona, S.; Jung, S. Origins and Tissue-context-dependent Fates of Blood Monocytes. Immunol. Cell Biol. 2009, 87, 30–38. [Google Scholar] [CrossRef]
  18. Sharifnia, T.; Antoun, J.; Verriere, T.G.C.; Suarez, G.; Wattacheril, J.; Wilson, K.T.; Peek, R.M.; Abumrad, N.N.; Flynn, C.R. Hepatic TLR4 Signaling in Obese NAFLD. Am. J. Physiol.-Gastrointest. Liver Physiol. 2015, 309, G270–G278. [Google Scholar] [CrossRef]
  19. Kanuri, G.; Ladurner, R.; Skibovskaya, J.; Spruss, A.; Königsrainer, A.; Bischoff, S.C.; Bergheim, I. Expression of toll-like receptors 1–5 but not TLR 6-10 is elevated in livers of patients with non-alcoholic fatty liver disease. Liver Int. 2015, 35, 562–568. [Google Scholar] [CrossRef]
  20. Hotamisligil, G.S. Inflammation, Metaflammation and Immunometabolic Disorders. Nature 2017, 542, 177–185. [Google Scholar] [CrossRef]
  21. Xu, G.-X.; Wei, S.; Yu, C.; Zhao, S.-Q.; Yang, W.-J.; Feng, Y.-H.; Pan, C.; Yang, K.-X.; Ma, Y. Activation of Kupffer Cells in NAFLD and NASH: Mechanisms and Therapeutic Interventions. Front. Cell Dev. Biol. 2023, 11, 1199519. [Google Scholar] [CrossRef]
  22. Zhou, Y.; Zhang, H.; Yao, Y.; Zhang, X.; Guan, Y.; Zheng, F. CD4+ T Cell Activation and Inflammation in NASH-Related Fibrosis. Front. Immunol. 2022, 13, 967410. [Google Scholar] [CrossRef]
  23. Matsuda, M.; Seki, E. The Liver Fibrosis Niche: Novel Insights into the Interplay between Fibrosis-Composing Mesenchymal Cells, Immune Cells, Endothelial Cells, and Extracellular Matrix. Food Chem. Toxicol. 2020, 143, 111556. [Google Scholar] [CrossRef]
  24. Ito, S.; Yukawa, T.; Uetake, S.; Yamauchi, M. Serum Intercellular Adhesion Molecule-1 in Patients with Nonalcoholic Steatohepatitis: Comparison with Alcoholic Hepatitis. Alcohol. Clin. Exp. Res. 2007, 31, S83–S87. [Google Scholar] [CrossRef]
  25. Heymann, F.; Peusquens, J.; Ludwig-Portugall, I.; Kohlhepp, M.; Ergen, C.; Niemietz, P.; Martin, C.; van Rooijen, N.; Ochando, J.C.; Randolph, G.J.; et al. Liver Inflammation Abrogates Immunological Tolerance Induced by Kupffer Cells. Hepatology 2015, 62, 279–291. [Google Scholar] [CrossRef]
  26. Jenne, C.N.; Kubes, P. Immune Surveillance by the Liver. Nat. Immunol. 2013, 14, 996–1006. [Google Scholar] [CrossRef]
  27. Radaeva, S.; Sun, R.; Jaruga, B.; Nguyen, V.T.; Tian, Z.; Gao, B. Natural Killer Cells Ameliorate Liver Fibrosis by Killing Activated Stellate Cells in NKG2D-Dependent and Tumor Necrosis Factor–Related Apoptosis-Inducing Ligand–Dependent Manners. Gastroenterology 2006, 130, 435–452. [Google Scholar] [CrossRef]
  28. Yang, L.; Pang, Y.; Moses, H.L. TGF-β and Immune Cells: An Important Regulatory Axis in the Tumor Microenvironment and Progression. Trends Immunol. 2010, 31, 220–227. [Google Scholar] [CrossRef]
  29. Bendelac, A.; Savage, P.B.; Teyton, L. The Biology of NKT Cells. Annu. Rev. Immunol. 2007, 25, 297–336. [Google Scholar] [CrossRef]
  30. Arrese, M.; Cabrera, D.; Kalergis, A.M.; Feldstein, A.E. Innate Immunity and Inflammation in NAFLD/NASH. Dig. Dis. Sci. 2016, 61, 1294–1303. [Google Scholar] [CrossRef]
  31. Kotas, M.E.; Lee, H.-Y.; Gillum, M.P.; Annicelli, C.; Guigni, B.A.; Shulman, G.I.; Medzhitov, R. Impact of CD1d Deficiency on Metabolism. PLoS ONE 2011, 6, e25478. [Google Scholar] [CrossRef]
  32. Tajiri, K.; Shimizu, Y. Role of NKT Cells in the Pathogenesis of NAFLD. Int. J. Hepatol. 2012, 2012, 850836. [Google Scholar] [CrossRef]
  33. Syn, W.-K.; Agboola, K.M.; Swiderska, M.; Michelotti, G.A.; Liaskou, E.; Pang, H.; Xie, G.; Philips, G.; Chan, I.S.; Karaca, G.F.; et al. NKT-Associated Hedgehog and Osteopontin Drive Fibrogenesis in Non-Alcoholic Fatty Liver Disease. Gut 2012, 61, 1323–1329. [Google Scholar] [CrossRef]
  34. Lombardi, R.; Piciotti, R.; Dongiovanni, P.; Meroni, M.; Fargion, S.; Fracanzani, A.L. PD-1/PD-L1 Immuno-Mediated Therapy in NAFLD: Advantages and Obstacles in the Treatment of Advanced Disease. Int. J. Mol. Sci. 2022, 23, 2707. [Google Scholar] [CrossRef] [PubMed]
  35. Schuster, S.; Cabrera, D.; Arrese, M.; Feldstein, A.E. Triggering and Resolution of Inflammation in NASH. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 349–364. [Google Scholar] [CrossRef]
  36. Grohmann, M.; Wiede, F.; Dodd, G.T.; Gurzov, E.N.; Ooi, G.J.; Butt, T.; Rasmiena, A.A.; Kaur, S.; Gulati, T.; Goh, P.K.; et al. Obesity Drives STAT-1-Dependent NASH and STAT-3-Dependent HCC. Cell 2018, 175, 1289–1306.e20. [Google Scholar] [CrossRef]
  37. Lu, J.G.; Iyasu, A.; French, B.; Tillman, B.; French, S.W. Overexpression of MHCII by Hepatocytes in Alcoholic Hepatitis (AH) Compared to Non-Alcoholic Steatohepatitis (NASH) and Normal Controls. Alcohol 2020, 84, 27–32. [Google Scholar] [CrossRef]
  38. Jorch, S.K.; Kubes, P. An Emerging Role for Neutrophil Extracellular Traps in Noninfectious Disease. Nat. Med. 2017, 23, 279–287. [Google Scholar] [CrossRef] [PubMed]
  39. Peiseler, M.; Kubes, P. More Friend than Foe: The Emerging Role of Neutrophils in Tissue Repair. J. Clin. Investig. 2019, 129, 2629–2639. [Google Scholar] [CrossRef]
  40. Soehnlein, O.; Steffens, S.; Hidalgo, A.; Weber, C. Neutrophils as Protagonists and Targets in Chronic Inflammation. Nat. Rev. Immunol. 2017, 17, 248–261. [Google Scholar] [CrossRef] [PubMed]
  41. Arelaki, S.; Koletsa, T.; Sinakos, E.; Papadopoulos, V.; Arvanitakis, K.; Skendros, P.; Akriviadis, E.; Ritis, K.; Germanidis, G.; Hytiroglou, P. Neutrophil Extracellular Traps Enriched with IL-1β and IL-17A Participate in the Hepatic Inflammatory Process of Patients with Non-Alcoholic Steatohepatitis. Virchows Arch. 2022, 481, 455–465. [Google Scholar] [CrossRef] [PubMed]
  42. Krenkel, O.; Tacke, F. Liver Macrophages in Tissue Homeostasis and Disease. Nat. Rev. Immunol. 2017, 17, 306–321. [Google Scholar] [CrossRef]
  43. Maretti-Mira, A.C.; Golden-Mason, L.; Salomon, M.P.; Kaplan, M.J.; Rosen, H.R. Cholesterol-Induced M4-Like Macrophages Recruit Neutrophils and Induce NETosis. Front. Immunol. 2021, 12, 671073. [Google Scholar] [CrossRef] [PubMed]
  44. Zhou, Z.; Xu, M.-J.; Cai, Y.; Wang, W.; Jiang, J.X.; Varga, Z.V.; Feng, D.; Pacher, P.; Kunos, G.; Torok, N.J.; et al. Neutrophil–Hepatic Stellate Cell Interactions Promote Fibrosis in Experimental Steatohepatitis. Cell. Mol. Gastroenterol. Hepatol. 2018, 5, 399–413. [Google Scholar] [CrossRef]
  45. Horst, A.K.; Tiegs, G.; Diehl, L. Contribution of Macrophage Efferocytosis to Liver Homeostasis and Disease. Front. Immunol. 2019, 10, 2670. [Google Scholar] [CrossRef]
  46. Racanelli, V.; Rehermann, B. The Liver as an Immunological Organ. Hepatology 2006, 43, S54–S62. [Google Scholar] [CrossRef] [PubMed]
  47. Krenkel, O.; Puengel, T.; Govaere, O.; Abdallah, A.T.; Mossanen, J.C.; Kohlhepp, M.; Liepelt, A.; Lefebvre, E.; Luedde, T.; Hellerbrand, C.; et al. Therapeutic Inhibition of Inflammatory Monocyte Recruitment Reduces Steatohepatitis and Liver Fibrosis. Hepatology 2018, 67, 1270–1283. [Google Scholar] [CrossRef]
  48. Parola, M.; Pinzani, M. Liver Fibrosis: Pathophysiology, Pathogenetic Targets and Clinical Issues. Mol. Asp. Med. 2019, 65, 37–55. [Google Scholar] [CrossRef] [PubMed]
  49. Anstee, Q.M.; Reeves, H.L.; Kotsiliti, E.; Govaere, O.; Heikenwalder, M. From NASH to HCC: Current Concepts and Future Challenges. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 411–428. [Google Scholar] [CrossRef] [PubMed]
  50. Parthasarathy, G.; Revelo, X.; Malhi, H. Pathogenesis of Nonalcoholic Steatohepatitis: An Overview. Hepatol. Commun. 2020, 4, 478–492. [Google Scholar] [CrossRef]
  51. Haas, J.T.; Francque, S.; Staels, B. Pathophysiology and Mechanisms of Nonalcoholic Fatty Liver Disease. Annu. Rev. Physiol. 2016, 78, 181–205. [Google Scholar] [CrossRef]
  52. De Silva, N.S.; Klein, U. Dynamics of B Cells in Germinal Centres. Nat. Rev. Immunol. 2015, 15, 137–148. [Google Scholar] [CrossRef] [PubMed]
  53. Bruzzì, S.; Sutti, S.; Giudici, G.; Burlone, M.E.; Ramavath, N.N.; Toscani, A.; Bozzola, C.; Schneider, P.; Morello, E.; Parola, M.; et al. B2-Lymphocyte Responses to Oxidative Stress-Derived Antigens Contribute to the Evolution of Nonalcoholic Fatty Liver Disease (NAFLD). Free Radic. Biol. Med. 2018, 124, 249–259. [Google Scholar] [CrossRef] [PubMed]
  54. Barrow, F.; Khan, S.; Fredrickson, G.; Wang, H.; Dietsche, K.; Parthiban, P.; Robert, S.; Kaiser, T.; Winer, S.; Herman, A.; et al. Microbiota-Driven Activation of Intrahepatic B Cells Aggravates NASH Through Innate and Adaptive Signaling. Hepatology 2021, 74, 704–722. [Google Scholar] [CrossRef] [PubMed]
  55. Litinskiy, M.B.; Nardelli, B.; Hilbert, D.M.; He, B.; Schaffer, A.; Casali, P.; Cerutti, A. DCs Induce CD40-Independent Immunoglobulin Class Switching through BLyS and APRIL. Nat. Immunol. 2002, 3, 822–829. [Google Scholar] [CrossRef] [PubMed]
  56. Miyake, T.; Abe, M.; Tokumoto, Y.; Hirooka, M.; Furukawa, S.; Kumagi, T.; Hamada, M.; Kawasaki, K.; Tada, F.; Ueda, T.; et al. B Cell-Activating Factor Is Associated with the Histological Severity of Nonalcoholic Fatty Liver Disease. Hepatol. Int. 2013, 7, 539–547. [Google Scholar] [CrossRef] [PubMed]
  57. Shuai, Z.; Leung, M.W.; He, X.; Zhang, W.; Yang, G.; Leung, P.S.; Eric Gershwin, M. Adaptive Immunity in the Liver. Cell. Mol. Immunol. 2016, 13, 354–368. [Google Scholar] [CrossRef] [PubMed]
  58. Selmi, C.; Podda, M.; Gershwin, M.E. Old and Rising Stars in the Lymphoid Liver. Semin. Immunopathol. 2009, 31, 279–282. [Google Scholar] [CrossRef] [PubMed]
  59. Guidotti, L.G.; Chisari, F.V. Immunology and pathogenesis of viral hepatitis. Annu. Rev. Pathol. Mech. Dis. 2006, 1, 23–61. [Google Scholar] [CrossRef] [PubMed]
  60. Taubert, R.; Hardtke-Wolenski, M.; Noyan, F.; Wilms, A.; Baumann, A.K.; Schlue, J.; Olek, S.; Falk, C.S.; Manns, M.P.; Jaeckel, E. Intrahepatic Regulatory T Cells in Autoimmune Hepatitis Are Associated with Treatment Response and Depleted with Current Therapies. J. Hepatol. 2014, 61, 1106–1114. [Google Scholar] [CrossRef]
  61. Sutti, S.; Jindal, A.; Locatelli, I.; Vacchiano, M.; Gigliotti, L.; Bozzola, C.; Albano, E. Adaptive Immune Responses Triggered by Oxidative Stress Contribute to Hepatic Inflammation in NASH. Hepatology 2014, 59, 886–897. [Google Scholar] [CrossRef]
  62. Murphy, K.M.; Stockinger, B. Effector T Cell Plasticity: Flexibility in the Face of Changing Circumstances. Nat. Immunol. 2010, 11, 674–680. [Google Scholar] [CrossRef]
  63. Hirsova, P.; Bamidele, A.O.; Wang, H.; Povero, D.; Revelo, X.S. Emerging Roles of T Cells in the Pathogenesis of Nonalcoholic Steatohepatitis and Hepatocellular Carcinoma. Front. Endocrinol. 2021, 12, 760860. [Google Scholar] [CrossRef]
  64. Luo, X.-Y.; Takahara, T.; Kawai, K.; Fujino, M.; Sugiyama, T.; Tsuneyama, K.; Tsukada, K.; Nakae, S.; Zhong, L.; Li, X.-K. IFN-γ Deficiency Attenuates Hepatic Inflammation and Fibrosis in a Steatohepatitis Model Induced by a Methionine- and Choline-Deficient High-Fat Diet. Am. J. Physiol.-Gastrointest. Liver Physiol. 2013, 305, G891–G899. [Google Scholar] [CrossRef]
  65. Omenetti, S.; Bussi, C.; Metidji, A.; Iseppon, A.; Lee, S.; Tolaini, M.; Li, Y.; Kelly, G.; Chakravarty, P.; Shoaie, S.; et al. The Intestine Harbors Functionally Distinct Homeostatic Tissue-Resident and Inflammatory Th17 Cells. Immunity 2019, 51, 77–89.e6. [Google Scholar] [CrossRef]
  66. Xu, R.; Tao, A.; Zhang, S.; Zhang, M. Neutralization of Interleukin-17 Attenuates High Fat Diet-Induced Non-Alcoholic Fatty Liver Disease in Mice. Acta Biochim. Biophys. Sin. 2013, 45, 726–733. [Google Scholar] [CrossRef]
  67. Rau, M.; Schilling, A.-K.; Meertens, J.; Hering, I.; Weiss, J.; Jurowich, C.; Kudlich, T.; Hermanns, H.M.; Bantel, H.; Beyersdorf, N.; et al. Progression from Nonalcoholic Fatty Liver to Nonalcoholic Steatohepatitis Is Marked by a Higher Frequency of Th17 Cells in the Liver and an Increased Th17/Resting Regulatory T Cell Ratio in Peripheral Blood and in the Liver. J. Immunol. 2016, 196, 97–105. [Google Scholar] [CrossRef]
  68. Ma, H.-Y.; Yamamoto, G.; Xu, J.; Liu, X.; Karin, D.; Kim, J.Y.; Alexandrov, L.B.; Koyama, Y.; Nishio, T.; Benner, C.; et al. IL-17 Signaling in Steatotic Hepatocytes and Macrophages Promotes Hepatocellular Carcinoma in Alcohol-Related Liver Disease. J. Hepatol. 2020, 72, 946–959. [Google Scholar] [CrossRef]
  69. Wedemeyer, H.; He, X.-S.; Nascimbeni, M.; Davis, A.R.; Greenberg, H.B.; Hoofnagle, J.H.; Liang, T.J.; Alter, H.; Rehermann, B. Impaired Effector Function of Hepatitis C Virus-Specific CD8+ T Cells in Chronic Hepatitis C Virus Infection1. J. Immunol. 2002, 169, 3447–3458. [Google Scholar] [CrossRef]
  70. Kaech, S.M.; Cui, W. Transcriptional Control of Effector and Memory CD8+ T Cell Differentiation. Nat. Rev. Immunol. 2012, 12, 749–761. [Google Scholar] [CrossRef]
  71. Breuer, D.A.; Pacheco, M.C.; Washington, M.K.; Montgomery, S.A.; Hasty, A.H.; Kennedy, A.J. CD8+ T Cells Regulate Liver Injury in Obesity-Related Nonalcoholic Fatty Liver Disease. Am. J. Physiol.-Gastrointest. Liver Physiol. 2020, 318, G211–G224. [Google Scholar] [CrossRef]
  72. Ghazarian, M.; Revelo, X.S.; Nøhr, M.K.; Luck, H.; Zeng, K.; Lei, H.; Tsai, S.; Schroer, S.A.; Park, Y.J.; Chng, M.H.Y.; et al. Type I Interferon Responses Drive Intrahepatic T Cells to Promote Metabolic Syndrome. Sci. Immunol. 2017, 2, eaai7616. [Google Scholar] [CrossRef]
  73. Bhattacharjee, J.; Kirby, M.; Softic, S.; Miles, L.; Salazar-Gonzalez, R.-M.; Shivakumar, P.; Kohli, R. Hepatic Natural Killer T-Cell and CD8+ T-Cell Signatures in Mice with Nonalcoholic Steatohepatitis. Hepatol. Commun. 2017, 1, 299–310. [Google Scholar] [CrossRef]
  74. Hansel, C.; Erschfeld, S.; Baues, M.; Lammers, T.; Weiskirchen, R.; Trautwein, C.; Kroy, D.C.; Drescher, H.K. The Inhibitory T Cell Receptors PD1 and 2B4 Are Differentially Regulated on CD4 and CD8 T Cells in a Mouse Model of Non-Alcoholic Steatohepatitis. Front. Pharmacol. 2019, 10, 244. [Google Scholar] [CrossRef]
  75. Wu, Q.; Jiang, L.; Li, S.; He, Q.; Yang, B.; Cao, J. Small Molecule Inhibitors Targeting the PD-1/PD-L1 Signaling Pathway. Acta Pharmacol. Sin. 2021, 42, 1–9. [Google Scholar] [CrossRef]
  76. Arasanz, H.; Gato-Cañas, M.; Zuazo, M.; Ibañez-Vea, M.; Breckpot, K.; Kochan, G.; Escors, D. PD1 Signal Transduction Pathways in T Cells. Oncotarget 2017, 8, 51936–51945. [Google Scholar] [CrossRef]
  77. Francisco, L.M.; Sage, P.T.; Sharpe, A.H. The PD-1 Pathway in Tolerance and Autoimmunity. Immunol. Rev. 2010, 236, 219–242. [Google Scholar] [CrossRef]
  78. Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and Its Ligands in Tolerance and Immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef]
  79. Kinter, A.L.; Godbout, E.J.; McNally, J.P.; Sereti, I.; Roby, G.A.; O’Shea, M.A.; Fauci, A.S. The Common γ-Chain Cytokines IL-2, IL-7, IL-15, and IL-21 Induce the Expression of Programmed Death-1 and Its Ligands. J. Immunol. 2008, 181, 6738–6746. [Google Scholar] [CrossRef]
  80. Chamoto, K.; Chowdhury, P.S.; Kumar, A.; Sonomura, K.; Matsuda, F.; Fagarasan, S.; Honjo, T. Mitochondrial Activation Chemicals Synergize with Surface Receptor PD-1 Blockade for T Cell-Dependent Antitumor Activity. Proc. Natl. Acad. Sci. USA 2017, 114, E761–E770. [Google Scholar] [CrossRef]
  81. O’Sullivan, D.; Pearce, E.L. Targeting T Cell Metabolism for Therapy. Trends Immunol. 2015, 36, 71–80. [Google Scholar] [CrossRef]
  82. Patsoukis, N.; Bardhan, K.; Chatterjee, P.; Sari, D.; Liu, B.; Bell, L.N.; Karoly, E.D.; Freeman, G.J.; Petkova, V.; Seth, P.; et al. PD-1 Alters T-Cell Metabolic Reprogramming by Inhibiting Glycolysis and Promoting Lipolysis and Fatty Acid Oxidation. Nat. Commun. 2015, 6, 6692. [Google Scholar] [CrossRef]
  83. Chang, C.-H.; Qiu, J.; O’Sullivan, D.; Buck, M.D.; Noguchi, T.; Curtis, J.D.; Chen, Q.; Gindin, M.; Gubin, M.M.; van der Windt, G.J.W.; et al. Metabolic Competition in the Tumor Microenvironment Is a Driver of Cancer Progression. Cell 2015, 162, 1229–1241. [Google Scholar] [CrossRef] [PubMed]
  84. Agata, Y.; Kawasaki, A.; Nishimura, H.; Ishida, Y.; Tsubat, T.; Yagita, H.; Honjo, T. Expression of the PD-1 Antigen on the Surface of Stimulated Mouse T and B Lymphocytes. Int. Immunol. 1996, 8, 765–772. [Google Scholar] [CrossRef] [PubMed]
  85. Riley, J.L. PD-1 Signaling in Primary T Cells. Immunol. Rev. 2009, 229, 114–125. [Google Scholar] [CrossRef] [PubMed]
  86. Patsoukis, N.; Li, L.; Sari, D.; Petkova, V.; Boussiotis, V.A. PD-1 Increases PTEN Phosphatase Activity While Decreasing PTEN Protein Stability by Inhibiting Casein Kinase 2. Mol. Cell. Biol. 2013, 33, 3091–3098. [Google Scholar] [CrossRef] [PubMed]
  87. Egen, J.G.; Kuhns, M.S.; Allison, J.P. CTLA-4: New Insights into Its Biological Function and Use in Tumor Immunotherapy. Nat. Immunol. 2002, 3, 611–618. [Google Scholar] [CrossRef] [PubMed]
  88. Takahashi, T.; Tagami, T.; Yamazaki, S.; Uede, T.; Shimizu, J.; Sakaguchi, N.; Mak, T.W.; Sakaguchi, S. Immunologic Self-Tolerance Maintained by Cd25+Cd4+Regulatory T Cells Constitutively Expressing Cytotoxic T Lymphocyte–Associated Antigen 4. J. Exp. Med. 2000, 192, 303–310. [Google Scholar] [CrossRef] [PubMed]
  89. Chae, Y.K.; Arya, A.; Iams, W.; Cruz, M.R.; Chandra, S.; Choi, J.; Giles, F. Current Landscape and Future of Dual Anti-CTLA4 and PD-1/PD-L1 Blockade Immunotherapy in Cancer; Lessons Learned from Clinical Trials with Melanoma and Non-Small Cell Lung Cancer (NSCLC). J. Immunother. Cancer 2018, 6, 39. [Google Scholar] [CrossRef] [PubMed]
  90. Schweitzer, A.N.; Sharpe, A.H. Studies Using Antigen-Presenting Cells Lacking Expression of Both B7-1 (CD80) and B7-2 (CD86) Show Distinct Requirements for B7 Molecules During Priming Versus Restimulation of Th2 But Not Th1 Cytokine Production1. J. Immunol. 1998, 161, 2762–2771. [Google Scholar] [CrossRef]
  91. Sawada, K.; Chung, H.; Softic, S.; Moreno-Fernandez, M.E.; Divanovic, S. The Bidirectional Immune Crosstalk in Metabolic Dysfunction-Associated Steatotic Liver Disease. Cell Metab. 2023, 35, 1852–1871. [Google Scholar] [CrossRef]
  92. Sharpe, A.H.; Freeman, G.J. The B7–CD28 Superfamily. Nat. Rev. Immunol. 2002, 2, 116–126. [Google Scholar] [CrossRef] [PubMed]
  93. Tivol, E.A.; Borriello, F.; Schweitzer, A.N.; Lynch, W.P.; Bluestone, J.A.; Sharpe, A.H. Loss of CTLA-4 Leads to Massive Lymphoproliferation and Fatal Multiorgan Tissue Destruction, Revealing a Critical Negative Regulatory Role of CTLA-4. Immunity 1995, 3, 541–547. [Google Scholar] [CrossRef] [PubMed]
  94. Chatzigeorgiou, A.; Chung, K.-J.; Garcia-Martin, R.; Alexaki, V.-I.; Klotzsche-von Ameln, A.; Phieler, J.; Sprott, D.; Kanczkowski, W.; Tzanavari, T.; Bdeir, M.; et al. Dual Role of B7 Costimulation in Obesity-Related Nonalcoholic Steatohepatitis and Metabolic Dysregulation. Hepatology 2014, 60, 1196–1210. [Google Scholar] [CrossRef] [PubMed]
  95. Chambers, C.A.; Kuhns, M.S.; Egen, J.G.; Allison, J.P. CTLA-4-Mediated Inhibition in Regulation of T Cell Responses: Mechanisms and Manipulation in Tumor Immunotherapy. Annu. Rev. Immunol. 2001, 19, 565–594. [Google Scholar] [CrossRef] [PubMed]
  96. Pinto, E.; Meneghel, P.; Farinati, F.; Russo, F.P.; Pelizzaro, F.; Gambato, M. Efficacy of Immunotherapy in Hepatocellular Carcinoma: Does Liver Disease Etiology Have a Role? Dig. Liver Dis. 1590. [Google Scholar] [CrossRef]
  97. Curran, M.A.; Montalvo, W.; Yagita, H.; Allison, J.P. PD-1 and CTLA-4 Combination Blockade Expands Infiltrating T Cells and Reduces Regulatory T and Myeloid Cells within B16 Melanoma Tumors. Proc. Natl. Acad. Sci. USA 2010, 107, 4275–4280. [Google Scholar] [CrossRef] [PubMed]
  98. Sun, C.; Mezzadra, R.; Schumacher, T.N. Regulation and Function of the PD-L1 Checkpoint. Immunity 2018, 48, 434–452. [Google Scholar] [CrossRef] [PubMed]
  99. Chauvin, J.-M.; Zarour, H.M. TIGIT in Cancer Immunotherapy. J. Immunother. Cancer 2020, 8, e000957. [Google Scholar] [CrossRef]
  100. Tao, L.; Song, C.; Huo, C.; Sun, Y.; Zhang, C.; Li, X.; Yu, S.; Sun, M.; Jin, B.; Zhang, Z.; et al. Anti-CD155 and anti-CD112 monoclonal antibodies conjugated to a fluorescent mesoporous silica nanosensor encapsulating rhodamine 6G and fluorescein for sensitive detection of liver cancer cells. Analyst 2016, 141, 4933–4940. [Google Scholar] [CrossRef] [PubMed]
  101. Zhu, Y.; Paniccia, A.; Schulick, A.C.; Chen, W.; Koenig, M.R.; Byers, J.T.; Yao, S.; Bevers, S.; Edil, B.H. Identification of CD112R as a Novel Checkpoint for Human T Cells. J. Exp. Med. 2016, 213, 167–176. [Google Scholar] [CrossRef]
  102. Tahara-Hanaoka, S.; Shibuya, K.; Onoda, Y.; Zhang, H.; Yamazaki, S.; Miyamoto, A.; Honda, S.; Lanier, L.L.; Shibuya, A. Functional Characterization of DNAM-1 (CD226) Interaction with Its Ligands PVR (CD155) and Nectin-2 (PRR-2/CD112). Int. Immunol. 2004, 16, 533–538. [Google Scholar] [CrossRef]
  103. Chauvin, J.-M.; Pagliano, O.; Fourcade, J.; Sun, Z.; Wang, H.; Sander, C.; Kirkwood, J.M.; Chen, T.T.; Maurer, M.; Korman, A.J.; et al. TIGIT and PD-1 Impair Tumor Antigen–Specific CD8+ T Cells in Melanoma Patients. J. Clin. Investig. 2015, 125, 2046–2058. [Google Scholar] [CrossRef] [PubMed]
  104. Joller, N.; Hafler, J.P.; Brynedal, B.; Kassam, N.; Spoerl, S.; Levin, S.D.; Sharpe, A.H.; Kuchroo, V.K. Cutting Edge: TIGIT Has T Cell-Intrinsic Inhibitory Functions. J. Immunol. 2011, 186, 1338–1342. [Google Scholar] [CrossRef] [PubMed]
  105. Ramsbottom, K.M.; Hawkins, E.D.; Shimoni, R.; McGrath, M.; Chan, C.J.; Russell, S.M.; Smyth, M.J.; Oliaro, J. Cutting Edge: DNAX Accessory Molecule 1–Deficient CD8+ T Cells Display Immunological Synapse Defects That Impair Antitumor Immunity. J. Immunol. 2014, 192, 553–557. [Google Scholar] [CrossRef] [PubMed]
  106. Zhang, Q.; Bi, J.; Zheng, X.; Chen, Y.; Wang, H.; Wu, W.; Wang, Z.; Wu, Q.; Peng, H.; Wei, H.; et al. Blockade of the Checkpoint Receptor TIGIT Prevents NK Cell Exhaustion and Elicits Potent Anti-Tumor Immunity. Nat. Immunol. 2018, 19, 723–732. [Google Scholar] [CrossRef]
  107. Winer, S.; Chan, Y.; Paltser, G.; Truong, D.; Tsui, H.; Bahrami, J.; Dorfman, R.; Wang, Y.; Zielenski, J.; Mastronardi, F.; et al. Normalization of Obesity-Associated Insulin Resistance through Immunotherapy. Nat. Med. 2009, 15, 921–929. [Google Scholar] [CrossRef]
  108. Nishimura, S.; Manabe, I.; Nagai, R. Adipose Tissue Inflammation in Obesity and Metabolic Syndrome. Discov. Med. 2009, 8, 55–60. [Google Scholar] [PubMed]
  109. Deng, J.; Liu, S.; Zou, L.; Xu, C.; Geng, B.; Xu, G. Lipolysis Response to Endoplasmic Reticulum Stress in Adipose Cells *. J. Biol. Chem. 2012, 287, 6240–6249. [Google Scholar] [CrossRef] [PubMed]
  110. Liu, R.; Nikolajczyk, B.S. Tissue Immune Cells Fuel Obesity-Associated Inflammation in Adipose Tissue and Beyond. Front. Immunol. 2019, 10, 1587. [Google Scholar] [CrossRef]
  111. Eljaafari, A.; Pestel, J.; Le Magueresse-Battistoni, B.; Chanon, S.; Watson, J.; Robert, M.; Disse, E.; Vidal, H. Adipose-Tissue-Derived Mesenchymal Stem Cells Mediate PD-L1 Overexpression in the White Adipose Tissue of Obese Individuals, Resulting in T Cell Dysfunction. Cells 2021, 10, 2645. [Google Scholar] [CrossRef]
  112. Shirakawa, K.; Yan, X.; Shinmura, K.; Endo, J.; Kataoka, M.; Katsumata, Y.; Yamamoto, T.; Anzai, A.; Isobe, S.; Yoshida, N.; et al. Obesity Accelerates T Cell Senescence in Murine Visceral Adipose Tissue. J. Clin. Investig. 2016, 126, 4626–4639. [Google Scholar] [CrossRef]
  113. Oldenhove, G.; Boucquey, E.; Taquin, A.; Acolty, V.; Bonetti, L.; Ryffel, B.; Le Bert, M.; Englebert, K.; Boon, L.; Moser, M. PD-1 Is Involved in the Dysregulation of Type 2 Innate Lymphoid Cells in a Murine Model of Obesity. Cell Rep. 2018; 25, 2053–2060.e4. [Google Scholar] [CrossRef]
  114. Schwartz, C.; Schmidt, V.; Deinzer, A.; Hawerkamp, H.C.; Hams, E.; Bayerlein, J.; Röger, O.; Bailer, M.; Krautz, C.; El Gendy, A.; et al. Innate PD-L1 Limits T Cell–Mediated Adipose Tissue Inflammation and Ameliorates Diet-Induced Obesity. Sci. Transl. Med. 2022, 14, eabj6879. [Google Scholar] [CrossRef] [PubMed]
  115. Azzu, V.; Vacca, M.; Virtue, S.; Allison, M.; Vidal-Puig, A. Adipose Tissue-Liver Cross Talk in the Control of Whole-Body Metabolism: Implications in Nonalcoholic Fatty Liver Disease. Gastroenterology 2020, 158, 1899–1912. [Google Scholar] [CrossRef]
  116. Nov, O.; Shapiro, H.; Ovadia, H.; Tarnovscki, T.; Dvir, I.; Shemesh, E.; Kovsan, J.; Shelef, I.; Carmi, Y.; Voronov, E.; et al. Interleukin-1β Regulates Fat-Liver Crosstalk in Obesity by Auto-Paracrine Modulation of Adipose Tissue Inflammation and Expandability. PLoS ONE 2013, 8, e53626. [Google Scholar] [CrossRef] [PubMed]
  117. Pedoeem, A.; Azoulay-Alfaguter, I.; Strazza, M.; Silverman, G.J.; Mor, A. Programmed Death-1 Pathway in Cancer and Autoimmunity. Clin. Immunol. 2014, 153, 145–152. [Google Scholar] [CrossRef] [PubMed]
  118. Sun, P.; Jin, Q.; Nie, S.; Jia, S.; Li, Y.; Li, X.; Guo, F. Unlike PD-L1, PD-1 Is Downregulated on Partial Immune Cells in Type 2 Diabetes. J. Diabetes Res. 2019, 2019, 1–8. [Google Scholar] [CrossRef] [PubMed]
  119. Fujisawa, R.; Haseda, F.; Tsutsumi, C.; Hiromine, Y.; Noso, S.; Kawabata, Y.; Mitsui, S.; Terasaki, J.; Ikegami, H.; Imagawa, A.; et al. Low Programmed Cell Death-1 (PD-1) Expression in Peripheral CD4+ T Cells in Japanese Patients with Autoimmune Type 1 Diabetes. Clin. Exp. Immunol. 2015, 180, 452–457. [Google Scholar] [CrossRef] [PubMed]
  120. Yao, A.; Liu, F.; Chen, K.; Tang, L.; Liu, L.; Zhang, K.; Yu, C.; Bian, G.; Guo, H.; Zheng, J.; et al. Programmed Death 1 Deficiency Induces the Polarization of Macrophages/Microglia to the M1 Phenotype After Spinal Cord Injury in Mice. Neurotherapeutics 2014, 11, 636–650. [Google Scholar] [CrossRef] [PubMed]
  121. Piątkiewicz, P.; Bernat-Karpińska, M.; Miłek, T.; Rabijewski, M.; Rosiak, E. NK Cell Count and Glucotransporter 4 (GLUT4) Expression in Subjects with Type 2 Diabetes and Colon Cancer. Diabetol. Metab. Syndr. 2016, 8, 38. [Google Scholar] [CrossRef] [PubMed]
  122. Pfister, D.; Núñez, N.G.; Pinyol, R.; Govaere, O.; Pinter, M.; Szydlowska, M.; Gupta, R.; Qiu, M.; Deczkowska, A.; Weiner, A.; et al. NASH Limits Anti-Tumour Surveillance in Immunotherapy-Treated HCC. Nature 2021, 592, 450–456. [Google Scholar] [CrossRef]
  123. Vardhana, S.A.; Hwee, M.A.; Berisa, M.; Wells, D.K.; Yost, K.E.; King, B.; Smith, M.; Herrera, P.S.; Chang, H.Y.; Satpathy, A.T.; et al. Impaired Mitochondrial Oxidative Phosphorylation Limits the Self-Renewal of T Cells Exposed to Persistent Antigen. Nat. Immunol. 2020, 21, 1022–1033. [Google Scholar] [CrossRef]
  124. Hashemi, M.; Karami, S.; Sarabandi, S.; Moazeni-Roodi, A.; Małecki, A.; Ghavami, S.; Wiechec, E. Association between PD-1 and PD-L1 Polymorphisms and the Risk of Cancer: A Meta-Analysis of Case-Control Studies. Cancers 2019, 11, 1150. [Google Scholar] [CrossRef] [PubMed]
  125. Eldafashi, N.; Darlay, R.; Shukla, R.; McCain, M.V.; Watson, R.; Liu, Y.L.; McStraw, N.; Fathy, M.; Fawzy, M.A.; Zaki, M.Y.W.; et al. A PDCD1 Role in the Genetic Predisposition to NAFLD-HCC? Cancers 2021, 13, 1412. [Google Scholar] [CrossRef] [PubMed]
  126. Pipitone, R.M.; Malvestiti, F.; Pennisi, G.; Jamialahmadi, O.; Dongiovanni, P.; Bertolazzi, G.; Pihlajamäki, J.; Yki-Järvinen, H.; Vespasiani-Gentilucci, U.; Tavaglione, F.; et al. Programmed Cell Death 1 Genetic Variant and Liver Damage in Nonalcoholic Fatty Liver Disease. Liver Int. 2023, 43, 1761–1771. [Google Scholar] [CrossRef] [PubMed]
  127. Schöniger, S.; Jasani, B. The PD-1/PD-L1 Pathway: A Perspective on Comparative Immuno-Oncology. Animals 2022, 12, 2661. [Google Scholar] [CrossRef]
  128. Mao, T.; Yang, R.; Luo, Y.; He, K. Crucial Role of T Cells in NAFLD-Related Disease: A Review and Prospect. Front. Endocrinol. 2022, 13, 1051076. [Google Scholar] [CrossRef]
  129. Pennisi, G.; Celsa, C.; Spatola, F.; Dallio, M.; Federico, A.; Petta, S. Pharmacological Therapy of Non-Alcoholic Fatty Liver Disease: What Drugs Are Available Now and Future Perspectives. Int. J. Environ. Res. Public Health 2019, 16, 4334. [Google Scholar] [CrossRef] [PubMed]
  130. Wen, Y.; Lambrecht, J.; Ju, C.; Tacke, F. Hepatic Macrophages in Liver Homeostasis and Diseases-Diversity, Plasticity and Therapeutic Opportunities. Cell. Mol. Immunol. 2021, 18, 45–56. [Google Scholar] [CrossRef] [PubMed]
  131. Raeman, R.; Anania, F.A. Therapy for Steatohepatitis: Do Macrophages Hold the Clue? Hepatology 2018, 67, 1204–1206. [Google Scholar] [CrossRef]
  132. Lefebvre, E.; Moyle, G.; Reshef, R.; Richman, L.P.; Thompson, M.; Hong, F.; Chou, H.; Hashiguchi, T.; Plato, C.; Poulin, D.; et al. Antifibrotic Effects of the Dual CCR2/CCR5 Antagonist Cenicriviroc in Animal Models of Liver and Kidney Fibrosis. PLoS ONE 2016, 11, e0158156. [Google Scholar] [CrossRef]
  133. Ratziu, V.; Sanyal, A.; Harrison, S.A.; Wong, V.W.-S.; Francque, S.; Goodman, Z.; Aithal, G.P.; Kowdley, K.V.; Seyedkazemi, S.; Fischer, L.; et al. Cenicriviroc Treatment for Adults With Nonalcoholic Steatohepatitis and Fibrosis: Final Analysis of the Phase 2b CENTAUR Study. Hepatology 2020, 72, 892–905. [Google Scholar] [CrossRef]
  134. Kliewer, S.A.; Forman, B.M.; Blumberg, B.; Ong, E.S.; Borgmeyer, U.; Mangelsdorf, D.J.; Umesono, K.; Evans, R.M. Differential Expression and Activation of a Family of Murine Peroxisome Proliferator-Activated Receptors. Proc. Natl. Acad. Sci. USA 1994, 91, 7355–7359. [Google Scholar] [CrossRef] [PubMed]
  135. Ratziu, V.; Harrison, S.A.; Francque, S.; Bedossa, P.; Lehert, P.; Serfaty, L.; Romero-Gomez, M.; Boursier, J.; Abdelmalek, M.; Caldwell, S.; et al. Elafibranor, an Agonist of the Peroxisome Proliferator-Activated Receptor-α and -δ, Induces Resolution of Nonalcoholic Steatohepatitis Without Fibrosis Worsening. Gastroenterology 2016, 150, 1147–1159.e5. [Google Scholar] [CrossRef] [PubMed]
  136. Francque, S.M.; Bedossa, P.; Ratziu, V.; Anstee, Q.M.; Bugianesi, E.; Sanyal, A.J.; Loomba, R.; Harrison, S.A.; Balabanska, R.; Mateva, L.; et al. A Randomized, Controlled Trial of the Pan-PPAR Agonist Lanifibranor in NASH. N. Engl. J. Med. 2021, 385, 1547–1558. [Google Scholar] [CrossRef] [PubMed]
  137. Neuschwander-Tetri, B.A.; Loomba, R.; Sanyal, A.J.; Lavine, J.E.; Van Natta, M.L.; Abdelmalek, M.F.; Chalasani, N.; Dasarathy, S.; Diehl, A.M.; Hameed, B.; et al. Farnesoid X Nuclear Receptor Ligand Obeticholic Acid for Non-Cirrhotic, Non-Alcoholic Steatohepatitis (FLINT): A Multicentre, Randomised, Placebo-Controlled Trial. Lancet 2015, 385, 956–965. [Google Scholar] [CrossRef]
  138. Younossi, Z.M.; Ratziu, V.; Loomba, R.; Rinella, M.; Anstee, Q.M.; Goodman, Z.; Bedossa, P.; Geier, A.; Beckebaum, S.; Newsome, P.N.; et al. Obeticholic Acid for the Treatment of Non-Alcoholic Steatohepatitis: Interim Analysis from a Multicentre, Randomised, Placebo-Controlled Phase 3 Trial. Lancet 2019, 394, 2184–2196. [Google Scholar] [CrossRef] [PubMed]
  139. Anstee, Q.M.; Lucas, K.J.; Francque, S.; Abdelmalek, M.F.; Sanyal, A.J.; Ratziu, V.; Gadano, A.C.; Rinella, M.; Charlton, M.; Loomba, R.; et al. Tropifexor plus Cenicriviroc Combination versus Monotherapy in Nonalcoholic Steatohepatitis: Results from the Phase 2b TANDEM Study. Hepatology 2023, 78, 1223–1239. [Google Scholar] [CrossRef]
  140. Ilan, Y.; Shailubhai, K.; Sanyal, A. Immunotherapy with Oral Administration of Humanized Anti-CD3 Monoclonal Antibody: A Novel Gut-Immune System-Based Therapy for Metaflammation and NASH. Clin. Exp. Immunol. 2018, 193, 275–283. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of physiological liver immunity status. In the healthy liver, the immunological homeostasis is maintained through the balance between basal pro-inflammatory cytokines, such as IL-2, IL-7, IL-12, and anti-inflammatory cytokines, like IL-10 and IL-13, and by the expression on PD-L1.Parenchymal and nonparenchymal cells, such as hepatocytes, HSCs, monocytes, KCs, and LSECs, contribute to maintaining liver immunity homeostasis cross-talk with T Reg cells.
Figure 1. Schematic representation of physiological liver immunity status. In the healthy liver, the immunological homeostasis is maintained through the balance between basal pro-inflammatory cytokines, such as IL-2, IL-7, IL-12, and anti-inflammatory cytokines, like IL-10 and IL-13, and by the expression on PD-L1.Parenchymal and nonparenchymal cells, such as hepatocytes, HSCs, monocytes, KCs, and LSECs, contribute to maintaining liver immunity homeostasis cross-talk with T Reg cells.
Ijms 25 03671 g001
Figure 2. Schematic representation of MASLD pathogenesis. Type 2 diabetes and obesity in adipose tissue promote a low state of chronic inflammation and an excessive release of free fatty acids (FFAs). The excessive uptake from FFAs into the liver activates the de novo lipogenesis (DNL) to convert the excess of free fatty acids (FFAs) and glucose into triglycerides (TGs). The high lipid content accumulates into hepatocytes, resulting in innate immunity activation and toxic molecules production, such as ROS, cytokines, and Neutrophil Extracellular Traps (NETs). The continuous chronic inflammatory environment activates the adaptative immune response, inducing the increase in T Reg, T CD8+ cells, the polarization of macrophages into the M1 phenotype, and the production of inflammatory cytokines, such as IFN-γ, TNF-α, IL-1, IL-6, and IL-1β. This persistent chronic inflammation occurs in all stages of MASLD progression, mostly in NASH, and during the transition to HCC, this inflammatory chronic status is also responsible for T CD8+ cells differentiating into exhausted T lymphocytes.
Figure 2. Schematic representation of MASLD pathogenesis. Type 2 diabetes and obesity in adipose tissue promote a low state of chronic inflammation and an excessive release of free fatty acids (FFAs). The excessive uptake from FFAs into the liver activates the de novo lipogenesis (DNL) to convert the excess of free fatty acids (FFAs) and glucose into triglycerides (TGs). The high lipid content accumulates into hepatocytes, resulting in innate immunity activation and toxic molecules production, such as ROS, cytokines, and Neutrophil Extracellular Traps (NETs). The continuous chronic inflammatory environment activates the adaptative immune response, inducing the increase in T Reg, T CD8+ cells, the polarization of macrophages into the M1 phenotype, and the production of inflammatory cytokines, such as IFN-γ, TNF-α, IL-1, IL-6, and IL-1β. This persistent chronic inflammation occurs in all stages of MASLD progression, mostly in NASH, and during the transition to HCC, this inflammatory chronic status is also responsible for T CD8+ cells differentiating into exhausted T lymphocytes.
Ijms 25 03671 g002
Figure 3. Schematic representation of immunity cells in MASLD and their role in promoting liver injury, fibrosis, and transition in HCC. A complex network and interactions between the liver’s resident cells, such as hepatocytes, Kupffer cells, and resident macrophages; immunity cells, such as neutrophils and B cells, involved in innate immune response; B cells; Th17 cells; Th1 cells; CD8+ cells; T Reg cells; and exhausted CD8+ T cells involved in adaptative immune response. Exhausted CD8+ cells are crucial players in the transition of MASH to HCC.
Figure 3. Schematic representation of immunity cells in MASLD and their role in promoting liver injury, fibrosis, and transition in HCC. A complex network and interactions between the liver’s resident cells, such as hepatocytes, Kupffer cells, and resident macrophages; immunity cells, such as neutrophils and B cells, involved in innate immune response; B cells; Th17 cells; Th1 cells; CD8+ cells; T Reg cells; and exhausted CD8+ T cells involved in adaptative immune response. Exhausted CD8+ cells are crucial players in the transition of MASH to HCC.
Ijms 25 03671 g003
Figure 4. Schematic representation of PD-1 signaling. The interaction of PD-1 and PD-L1 or PD-L2 leads to tyrosine phosphorylation (P) of the immunoreceptor tyrosine-based inhibitory motif (ITIM) and immunoreceptor tyrosine-based switch Motif (ITSM). This phosphorylation recruits SHP-1 (Src homology phosphatase 1) and SHP-2 (Src homology phosphatase 2) that act on proximal signaling molecules and reinforce PTEN expression, thus effectively reducing the PI3K (phosphoinositide-3-kinase) and RAS (rat sarcoma) pathways. PD-1 signaling may result in decreased T-cell proliferation, survival, protein synthesis, and cytokine production. (Red arrows describe the inhibitory effect in PD-1-mediated signaling, whereas the blue arrow indicates the consequence of antagonist signaling CD80/CD28 of PD-1/PD-L1).
Figure 4. Schematic representation of PD-1 signaling. The interaction of PD-1 and PD-L1 or PD-L2 leads to tyrosine phosphorylation (P) of the immunoreceptor tyrosine-based inhibitory motif (ITIM) and immunoreceptor tyrosine-based switch Motif (ITSM). This phosphorylation recruits SHP-1 (Src homology phosphatase 1) and SHP-2 (Src homology phosphatase 2) that act on proximal signaling molecules and reinforce PTEN expression, thus effectively reducing the PI3K (phosphoinositide-3-kinase) and RAS (rat sarcoma) pathways. PD-1 signaling may result in decreased T-cell proliferation, survival, protein synthesis, and cytokine production. (Red arrows describe the inhibitory effect in PD-1-mediated signaling, whereas the blue arrow indicates the consequence of antagonist signaling CD80/CD28 of PD-1/PD-L1).
Ijms 25 03671 g004
Figure 5. Schematic representation of MASLD pathogenesis and HCC transition. The accumulation of exhausted cytotoxic T cells is involved in the progression of liver disease. T lymphocyte activation and persistent expression of PD-1 are associated with the loss of T cell activity. CD8+ PD-1+CXCR6 lymphocytes impair immune surveillance, contributing to tissue damage and progression. Several single-nucleotide polymorphisms have been correlated with higher PD-1 expression in T lymphocytes.
Figure 5. Schematic representation of MASLD pathogenesis and HCC transition. The accumulation of exhausted cytotoxic T cells is involved in the progression of liver disease. T lymphocyte activation and persistent expression of PD-1 are associated with the loss of T cell activity. CD8+ PD-1+CXCR6 lymphocytes impair immune surveillance, contributing to tissue damage and progression. Several single-nucleotide polymorphisms have been correlated with higher PD-1 expression in T lymphocytes.
Ijms 25 03671 g005
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pipitone, R.M.; Lupo, G.; Zito, R.; Javed, A.; Petta, S.; Pennisi, G.; Grimaudo, S. The PD-1/PD-L1 Axis in the Biology of MASLD. Int. J. Mol. Sci. 2024, 25, 3671. https://doi.org/10.3390/ijms25073671

AMA Style

Pipitone RM, Lupo G, Zito R, Javed A, Petta S, Pennisi G, Grimaudo S. The PD-1/PD-L1 Axis in the Biology of MASLD. International Journal of Molecular Sciences. 2024; 25(7):3671. https://doi.org/10.3390/ijms25073671

Chicago/Turabian Style

Pipitone, Rosaria Maria, Giulia Lupo, Rossella Zito, Ayesha Javed, Salvatore Petta, Grazia Pennisi, and Stefania Grimaudo. 2024. "The PD-1/PD-L1 Axis in the Biology of MASLD" International Journal of Molecular Sciences 25, no. 7: 3671. https://doi.org/10.3390/ijms25073671

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop