Minireviews Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Transplant. Sep 18, 2025; 15(3): 103163
Published online Sep 18, 2025. doi: 10.5500/wjt.v15.i3.103163
Role of autophagy in rejection after solid organ transplantation: A systematic review of the literature
Shu-Min Jiang, Zi-Lin Wang, Zhi-Wei Chen, Zhi-Long Liu, Qiang Li, Xiao-Long Chen, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
Xue-Jiao Li, Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, China
ORCID number: Xiao-Long Chen (0000-0003-1566-6975).
Co-first authors: Shu-Min Jiang and Xue-Jiao Li.
Co-corresponding authors: Qiang Li and Xiao-Long Chen.
Author contributions: Jiang SM, Li XJ, and Chen XL were responsible for drafting the manuscript; Jiang SM and Li XJ contributed equally to this paper and are the co-first authors of this manuscript; Wang ZL, Chen ZW, and Liu ZL were responsible for data acquisition; Li Q and Chen XL were responsible for submitting the version for final approval, they contributed equally to this paper and are the co-corresponding authors of this manuscript; Chen XL was responsible for conception and design, manuscript revision, and funding; and all authors thoroughly reviewed and endorsed the final manuscript.
Supported by the National Natural Science Foundation of China, No. 82100691; and China Postdoctoral Science Foundation, No. 2021M693631.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Xiao-Long Chen, Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, No. 613 Huangpu Avenue, Guangzhou 510630, Guangdong Province, China. chenxl0406@jnu.edu.cn
Received: November 18, 2024
Revised: January 21, 2025
Accepted: February 6, 2025
Published online: September 18, 2025
Processing time: 152 Days and 4.6 Hours

Abstract

Organ transplantation has long been recognized as an effective treatment for end-stage organ failure, metabolic diseases, and malignant tumors. However, graft rejection caused by major histocompatibility complex mismatch remains a significant challenge. While modern immunosuppressants have made significant strides in reducing the incidence and risk of rejection, they have not been able to eliminate it completely. The intricate mechanisms underlying transplant rejection have been the subject of intense investigation by transplant immunologists. Among these factors, autophagy has emerged as a key player. Autophagy is an evolutionarily conserved mechanism in eukaryotic cells that mediates autophagocytosis and cellular protection. This process is regulated by autophagy-related genes and their encoded protein families, which maintain the material and energetic balance within cells. Additionally, autophagy has been reported to play crucial roles in the development, maturation, differentiation, and responses of immune cells. In the complex immune environment following transplantation, the role and mechanisms of autophagy are gradually being revealed. In this review, we aim to explore the current understanding of the role of autophagy in solid organ rejection after transplantation. Furthermore, we delve into the therapeutic advancements achieved by targeting autophagy involved in the rejection process.

Key Words: Autophagy; Organ transplantation; Graft rejection; Immunity; Immune modulation

Core Tip: The intricate mechanisms underlying transplant rejection have been the subject of intense investigation by transplant immunologists. As an evolutionarily conserved self-regulatory protective mechanism in eukaryotic cells, autophagy also plays a crucial role in the development, maturation, differentiation and response of immune cells. Our article reviews the role and mechanisms of autophagy of different types of immune cells in the rejection after solid organ transplantation, so as to provide a theoretical basis for better understanding the profound connotation of graft rejection and potential treatment approaches by targeting autophagy in the future.



INTRODUCTION

Organ transplantation, a life-saving procedure, has been extensively utilized globally since its initial success in the 1950s[1-3]. Owing to continual advancements in medical technology, the success rates of organ transplants have notably increased, offering renewed hope to numerous patients suffering from end-stage organ failure. Nevertheless, immune rejection remains a significant hurdle that impacts long-term graft survival and patient quality of life, despite remarkable progress in transplantation techniques. Immune rejection occurs when the recipient’s immune system attacks the graft’s alloantigens, potentially leading to graft dysfunction, severe complications, and even life-threatening situations for the patient[4]. While clinically administered immunosuppressants can mitigate rejection to some extent, their long-term use is associated with numerous side effects and cannot fully prevent rejection[5]. Consequently, effective rejection inhibition and enhanced long-term graft survival rates remain pivotal and challenging aspects in the transplantation field.

In recent years, autophagy, an important cellular self-regulatory mechanism, has gradually been recognized for its role in the immune system. Autophagy is not only involved in the degradation and recycling of intracellular substances but also plays an important role in the development, differentiation and function of immune cells[6-8]. In particular, in organ transplant rejection, the regulatory effect of autophagy on immune cells may have a profound impact on graft survival and patient prognosis. In this review, we discuss the research progress on autophagy and its molecular mechanism in organ transplantation rejection and its potential therapeutic value to provide new ideas and methods for the clinical practice of organ transplantation.

BASIC CONCEPTS AND MECHANISMS OF AUTOPHAGY
Definition and classification of autophagy

Autophagy is a highly conserved biological process in eukaryotic cells that involves the degradation and recycling of senescent, damaged or redundant organelles and proteins through lysosomes to maintain cell homeostasis[9-11]. Autophagy can be classified into macroautophagy, microautophagy and chaperone-mediated autophagy according to the pathway of substrate entry into lysosomes. Among them, macroautophagy is the most widely studied, and autophagy is usually referred to as macroautophagy.

Processes and regulation of autophagy

The process of autophagy includes the formation of autophagic vesicles, the fusion of autophagosomes and lysosomes, and the degradation of substances in autophagosomes[12]. The regulatory mechanisms of autophagy are complex and diverse and involve a variety of signaling pathways and molecules. Under conditions such as nutrient deficiency, oxidative stress, and pathogen infection, the level of autophagy in cells significantly increases in response to external stimuli. Autophagy is regulated mainly by proteins encoded by autophagy-related genes (ATGs), which play key roles in the formation, fusion and degradation of autophagic vesicles[13]. The molecular regulatory mechanisms of autophagy involve multiple key proteins and signaling pathways that play important roles in the initiation, expansion, and completion of autophagy. The core regulatory proteins involved in autophagy include members of the ATG-related proteins family, such as ATG 5, ATG 7, ATG 12, and microtubule-associated protein 1 light chain 3 (LC3), which can form two types of ubiquitin-like proteins and link systems: (1) The ATG 12-ATG 5-ATG 16 system, which is activated by ATG 7 (E1-like kinase), and is responsible for the excitation and positioning of the LC3 link system by participating in the expansion of autophagosomes; and (2) The LC3 link system, in which the LC3 precursor is modified by ATG 4 to form LC3-I, and its lipid-binding site - lipid phosphatidylethanolamine (PE) is bound by ATG 7 and ATG 3 to form a lipid-soluble LC3-II-PE. Since LC3-II can bind to the autophagosome membrane, it often serves as a marker for autophagosome formation[14].

During the induction phase of autophagy, the ATG 1/ unc-51-like kinase 1 (ULK 1) kinase complex plays a major role. ATG 1 is a protein serine/threonine kinase, but its own kinase activity does not affect the process of autophagy. ATG 1 can bind to ATG 13 (highly phosphorylated), activate the phosphatase protein phosphatase 2A to dephosphorylate ATG 13, and increase the binding to ATG 1. The ATG 1-ATG 13 complex binds to ATG 17-29-31 to initiate autophagy. In mammals, ULK 1/2, as a homolog of ATG 1, forms a ULK 1/2 complex with ATG 13 and focal adhesion kinase family interacting protein of 200 kD, which is a mammalian target of rapamycin (mTOR). When stimulated by energy deprivation or various factors, mTOR activity is reduced, which results in the activation of adenosine phosphate-activated protein kinase, the activation and phosphorylation of the ULK 1/2 complex, and the induction of phagosome nucleation. The precursor nucleation phase is mainly mediated by the class III phosphoinositide 3-kinase complex, which includes vacuolar sorting protein 34, beclin1 (ATG 6 homolog) and ATG 14. This complex mediates phagocytic bubble nucleation while recruiting the ATG 12-ATG 5-ATG 16 complex as well as LC3 to play a ubiquitin-like role in promoting the elongation of the phagocytic bubble membrane. With the expansion and closure of autophagosomes, they gradually mature. Under the action of the cytoskeleton and related motor proteins, tethering factors and soluble N-ethyl maleimide sensitive factor attachment protein receptor-related proteins promote the fusion of lysosomes to form autophagosomes. Their inner membrane and contents are degraded by lipases, proteases and proteases in lysosomes into small molecules for cell utilization[15].

ORGAN TRANSPLANTATION REJECTION AND IMMUNE TOLERANCE
Types and mechanisms of organ transplant rejection

Organ transplant rejection can be categorized into hyperacute, acute, and chronic rejection[16]. Hyperacute rejection, which occurs within minutes to hours post-transplantation, is caused primarily by ABO blood group incompatibilities or preexisting antibodies[17]. Acute rejection, the most prevalent type, typically manifests within weeks to months following transplantation. This rejection is predominantly mediated by T lymphocytes responding to the recognition of allotypic antigens on the graft[18,19]. Chronic rejection, a delayed form, often arises within months or years after the procedure. It involves a complex interplay between immune and nonimmune cells, leading to a gradual decline in graft function[20,21].

Immunological mechanisms of graft rejection

The immune response underlying transplant rejection involves the activation and proliferation of various immune cells, including T lymphocytes, B lymphocytes, macrophages, and dendritic cells (DCs), as well as the secretion and regulatory actions of cytokines. T lymphocytes play a pivotal role in recognizing and attacking graft alloantigens. B lymphocytes contribute by producing antibodies, whereas macrophages and DCs, as antigen-presenting cells (APCs), initiate immune responses by capturing, processing, and presenting antigens to T lymphocytes. Cytokine secretion and regulation influence the type and intensity of the immune response.

Rejection is initiated by the recognition of graft antigens. Donor-derived APCs, particularly the DCs remaining in the graft, present the intact donor peptide-major histocompatibility complex (pMHC) without antigen processing to the recipient’s T lymphocytes via direct recognition[22]. Recipient APCs repeat this process, processing and presenting antigens to naive T lymphocytes through indirect recognition mechanisms[23]. Additionally, a semi-indirect recognition pathway exists in which recipient APCs present intact donor alloantigen pMHC complexes transferred from donor APC to recipient T cells by exosome or other means[24]. Among these pathways, direct recognition is considered the most critical. The characteristics of different recognition ways are shown in Figure 1.

Figure 1
Figure 1 The characteristics of different recognition ways in organ transplantation. Direct recognition: Recipient T cells directly recognize intact alloantigen peptide-major histocompatibility complex presented by the donor antigen presenting cell (APC) without antigen processing; Indirect recognition: Recipient T cells recognize the donor alloantigen-self major histocompatibility complex presented by recipient APC processing; Semi-direct recognition: Recipient T cells recognize intact donor alloantigen peptide-major histocompatibility complex on the surface of recipient APC that transferred from donor APC by exosome or other means. MHC: Major histocompatibility complex; APC: Antigen presenting cell; TCR: T-cell receptor; p-MHC: Peptide-major histocompatibility complex.

The recognition of transplantation antigens provides the initial signal for T cell activation, whereas the interaction of the costimulatory molecules CD80/86 and CD40 on APCs with the T cell surface receptor CD28 and CD40 L provides the second signal necessary for lymphocyte activation, enabling the full activation of reactive T lymphocytes. Besides, inflammation cytokine signals such as interleukin (IL)-2/IL-2R further promote the activation T cells. These signals are transmitted intracellularly via the calcium - calcineurin pathway, mTOR signaling, and nuclear factor of activated T cells (NF-AT) pathway, activating transcription factors that produce IL-2 and other proinflammatory cytokines and promote T cell proliferation. This stimulates lymphocyte proliferation and differentiation into effector T and B lymphocytes[25-27]. The signaling pathway of activation and proliferation of T cells after recognizing alloantigen is shown in Figure 2.

Figure 2
Figure 2 The signaling pathway of activation and proliferation of T cells after recognizing alloantigen. Alloantigen recognition provides the initial signal for T cell activation, then the interaction of the costimulatory molecules CD80/86 and CD40 on antigen presenting cell and B cell with the T cell surface receptor CD28 and CD40 L provides the second signal, enabling the full activation of reactive T lymphocytes. These signals then transmitted intracellularly activating the calcium-calcineurin pathway and then promoting dephosphorylation of nuclear factor of activated T cells and its nuclear translocation, resulting in activating transcription factors that produce interleukin (IL)-2 and other proinflammatory cytokines. Inflammation cytokine IL-2 engagement with IL-2R activating mammalian target of rapamycin complex 1 and then cyclin D1/CDK4 signaling, thus promote cell cycle from G1 to S phase and ultimately facilitate proliferation of T cells. APC: Antigen presenting cell; MHC: Major histocompatibility complex; NF-AT: Nuclear factor of activated T cells; TCR: T-cell receptor; mTOR: Mammalian target of rapamycin; IL: Interleukin; PRAS40: Proline-rich Akt substrate of 40 kDa; mLST80: Mammalian lethal with SEC13 protein 8.
Induction and maintenance of immune tolerance

Immune tolerance refers to the immune system’s lack of response, or production of a minimal immune reaction, to a specific antigen[28]. This serves to preserve the stability of the body’s internal environment. In the context of organ transplantation, inducing immune tolerance plays a crucial role in minimizing the need for immunosuppressive drugs, mitigating the chances of rejection, and enhancing graft survival. Nevertheless, the precise mechanism underlying immune tolerance induction remains elusive and warrants further exploration.

Role of autophagy in organ transplant rejection

Autophagy can regulate the activity and function of a variety of immune cells. For example, autophagy can regulate the maturation and function of DCs, affecting their antigen presentation and immune stimulation ability[29]. Autophagy can also regulate the metabolic state and signaling pathways of lymphocytes, affecting their proliferation, differentiation and survival[30,31]. In addition, autophagy can affect the activity and function of innate immune cells, such as macrophages and natural killer cells, and regulate their ability to phagocytose, kill and secrete cytokines[32]. Here, we compiled the autophagy status of different immune cells from literature on various organ transplantation models, as well as the impact of manipulating autophagy on immune cells and transplantation rejection outcomes (Table 1).

Table 1 Role of autophagy in different immune cells in organ transplantation rejection.
Cell type
Transplant model
Autophagy level in transplantation
Autophagy manipulating
Autophagy level after manipulating
Functional outcome after autophagy manipulating
Ref.
T cellHeart, miceN/ABECN1 knock down N/AIncreased alloreactivity of T cells; Enhanced graft rejectionVerghese et al[37], 2014
Liver, rats3-methyladenineInhibited survival and function of T cells; prolonged allograft survivalChen et al[38], 2019
Skin and heart, miceN/AChloroquineN/AReduced infiltration of T cells; prolonged allograft survivalCui et al[40], 2020
Liver, ratsJNK inhibitor SP600125Inhibited survival and function of T cells; prolonged allograft survivalWang et al[39], 2024
B cellHeart, miceATG7 knock downInhibited secondary alloantibody responses, decreased alloreactive memory B cellsFribourg et al[45], 2018
DCSkin, miceN/ASorafenibInhibited viability, maturation of dendritic cells; prolonged allograft survivalLin et al[58], 2013
TregHeart, miceThe miR-146a knock downEnhanced inhibitory effects of Tregs; prolonged allograft survivalLu et al[65], 2021
Heart, micelncRNA PVT1 overexpressionEnhanced inhibitory effects of Tregs; prolonged allograft survivalLu et al[66], 2021
MDSCHeart, miceN/AmTOR conditional knockoutPromoted differentiation, immunosuppressive function of MDSCs; prolonged allograft survivalLi et al[68], 2021
MacrophageCorneal, ratsN/AKaempferolReduced M1 polarization, inflammasome activation of macrophage; alleviated graft rejectionTian et al[73], 2021
Effects of autophagy on T lymphocytes

Autophagy is involved in the differentiation and functional development of T lymphocytes. Although bone marrow cells can differentiate in the absence of autophagy, severe bone marrow hyperplasia can occur, and the lack of autophagy in mouse hematopoietic progenitor cells severely impairs the differentiation of the lymphoid lineage[33]. Thymocyte development is more dependent on functional autophagy, and in the absence of autophagy to remove damaged mitochondria (mitophagy), the number of naïve T cells is greatly reduced[8]. Autophagy also plays a crucial role in the functional development of mature T cells. The survival of T cells during proliferation depends on autophagy, and a lack of autophagy leads to mitochondrial dysfunction and promotes the aging and apoptosis of T cells[34]. In addition, a lack of autophagy leads to impaired memory formation in CD8+ T cells, whereas the promotion of autophagy helps to restore T cell memory responses[35,36].

Graft antigen-reactive T cells are undoubtedly the key actors leading to organ rejection injury. Although the important role of autophagy in the biological function of T cells has been described above, the role of autophagy in T cells in the microenvironment of transplantation is still controversial. An earlier study in a mouse heart transplantation model suggested that autophagy is necessary to promote effector T cell death during transplantation tolerance induction, and autophagy-deficient beclin-1 heterozygous effector T cells adoptively transferred to allogeneic recipients exhibited increased proliferation, reduced cell death, and increased Interferon-γ production[37]. In the rat liver transplantation rejection model, we confirmed that autophagy in CD8+ T cells is a key factor mediating liver transplantation rejection[38]. The underlying mechanism is that upregulated jun N-terminal kinases signaling promotes beclin 1/ B-cell lymphoma-2 complex depolymerization and upregulates beclin 1 expression, which activates autophagy to promote the survival and proliferation of CD8+ T cells. Pharmacological inhibition of autophagy with the inhibitor 3-methyladenine (3-MA) or inhibition of jun N-terminal kinases signaling significantly prolonged receptor survival[39]. Similarly, a study involving mouse skin and heart transplant models revealed that the administration of chloroquine, another autophagy inhibitor, inhibited the activation and function of murine cardiac alloreactive T cells by increasing cytotoxic-T-lymphocyte-associated antigen 4 expression during T-cell activation in vitro and in vivo[40]. However, in graft-versus-host disease (GVHD), which has immunological mechanisms similar to those of rejection, the function of autophagy in T cells is again complicated. Oravecz-Wilson et al[41] reported that allogeneic stimulation can promote autophagy in T cells both in vitro and in vivo, but ATG5 deficiency in donor T cells attenuates the induction of autophagy, decreases proliferation, and promotes an increase in effector cytokines. This uncoupling of proliferation and the effector response ultimately alleviates GVHD. Although the reason for these differences is not clear, it may be partly due to the differences in the organ transplant models involved, as the immune microenvironment itself varies greatly from one transplant model to another. The difference in autophagy research methods is also one of the reasons. The detection of autophagic flow and autophagic flux is considered the gold standard for detecting the level of autophagy, but it is very difficult for immune cells to operate, and simply using flow cytometry, immunohistochemistry, and western blotting to detect the expression of autophagy-related proteins may mask true autophagy. The abovementioned factors ultimately limit the unity of the conclusions of different researchers on the role of T cell autophagy in organ transplantation rejection.

Effects of autophagy on B lymphocytes

In the past, B cells were considered participants in graft rejection rather than initiators of it. B cells have been known to participate in graft rejection by providing a second activation signal, secreting cytokines, and acting as APCs to activate T cells[42]. Recently, B cells have been shown to secrete antibodies to induce antibody-mediated rejection (AMR), which has attracted increasing attention in organ transplantation. AMR is mediated primarily by donor-specific antibodies that usually target both human leukocyte antigen (HLA) and non-HLA antigens in the donor organ, leading to complement activation, inflammatory responses, and tissue damage. The recipient’s preexisting anti-donor HLA antibodies or those produced by the recipient’s B cells activated by transplantation antigens after transplantation damage the graft by activating complement and antibody-dependent cellular cytotoxicity, resulting in rejection[43]. Notably, AMR is an important mechanism mediating rejection after kidney and heart transplantation, but it is relatively rare in liver transplantation[44].

In addition to its role in T cells, autophagy plays an important role in regulating B cell function and survival; however, other detailed reviews are lacking. We discuss the role and mechanism of B cell autophagy in the microenvironment of transplantation. In a mouse model of heart transplantation, Fribourg et al[45] reported that autophagy is required for memory B cell function and secondary alloantibody responses and that the inhibition of autophagy with the inhibitor 3-MA prevents memory donor-specific antigen responses to alloantigens. A renal transplant clinical cohort study revealed that the expression of ATGs was significantly associated with the development of AMR[46]. Zhang et al[47] reported that the levels of the autophagy-related protein ATG12 and autophagy were increased in the B cells of patients with AMR after kidney transplantation. Further application of miR-107 may regulate autophagy by targeting ATG12 to reduce the secretion of IgG and IgM antibodies by the B lymphocytes of transplant recipients. These studies suggest that autophagy in B cells promotes rejection by regulating function and memory formation.

Effects of autophagy on DCs

DCs are important APCs in the immune system that can activate T lymphocytes and trigger immune responses, thus playing an important role in organ transplantation rejection. Autophagy plays an important role in the maturation and function of DCs[48]. Autophagy can regulate the metabolic state and functional activity of DCs, thereby affecting their ability to take up, process and present antigens. During endogenous antigen presentation, autophagy contributes to the degradation of endogenous antigens into peptides. These peptides can then be transported to the endoplasmic reticulum by transporters associated with antigen processing, where they are further processed and bound to major histocompatibility complex (MHC) I molecules to form peptide-MHCI complexes and activate CD8+ T cells[49,50]. In addition, autophagy is required for MHC II molecules to present antigens in vivo, and dendritic cell-mediated LC3-PE-coupled autophagy can promote MHC II molecules to present foreign antigens to CD4+ T cells[51], which further expands the role of autophagy in antigen presentation. In contrast to its role in antigen presentation, most of the current literature suggests that autophagy has an inhibitory role in the immunogenic maturation of DCs[52-54]. Immature DCs, which have low expression of MHC II class molecules and CD40 (-), CD80 (-) and CD86 (-) stimulating molecules and a reduced ability to stimulate the T cell response, can induce antigen-specific T cell apoptosis or anergy[55,56]. In addition, autophagy is also thought to inhibit the migration of DCs to lymph nodes, which may be related to the regulatory effect of autophagy on cytoskeletal proteins[57].

The paradoxical role of autophagy in antigen presentation and immunogenic maturation in DCs undoubtedly complicates its role in organ transplant rejection. A study using a mouse skin graft model revealed that sorafenib induced autophagy in DCs and inhibited their maturation and survival, thereby reducing graft rejection injury and prolonging skin graft survival[58]. Unfortunately, very few studies have investigated the role of DC autophagy in transplant rejection. Some researchers have explored the role of dendritic cell autophagy in the graft-versus-host reaction in bone marrow hematopoietic stem cell transplantation, which may provide some insight. A study of DCs after hematopoietic cell transplantation revealed that complement C3aR/C5aR signaling promoted host DC survival and activation by inhibiting lethal mitophagy, whereas blocking C3aR/C5aR activation significantly improved GVHD outcomes after hematopoietic cell transplantation (HCT) by enhancing mitophagy in recipient DCs[59]. Undoubtedly, regardless of the mechanism, these studies suggest that autophagy may play a negative regulatory role in DCs and may be beneficial for the amelioration of rejection.

Effects of autophagy on regulatory cells

In the graft microenvironment, the major regulatory cell populations include regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), the latter of which are a heterogeneous group of bone marrow-derived cells that are precursors of DCs, macrophages, and/or granulocytes[60-62]. The regulatory T cell population can inhibit immune responses, not only through the cytotoxicity of natural killer cells and natural killer T cells, but also through the immune response mediated by CD4+ and CD8+ T cells. Therefore, Tregs are considered to negatively regulate rejection and promote the development of immune tolerance.

Tregs are a subset of T cells with immunosuppressive functions that play important roles in maintaining immune homeostasis and preventing autoimmune diseases. Autophagy plays an important role in the function and stability of Tregs. Autophagy is considered to maintain the metabolic homeostasis and functional activity of Tregs, thereby enhancing their immunosuppressive capacity. In addition, autophagy can promote the proliferation and differentiation of Tregs[63,64]. In the transplant microenvironment, the regulatory role of autophagy in Tregs was also revealed. A mouse heart transplantation model revealed that miR-146a knockdown enhanced Treg autophagy and function, and autophagy-enhanced Tregs exhibited significant immunosuppressive functions and therapeutic efficacy for graft rejection[65]. In another heart transplantation study, the mechanism of miR-146a-induced Treg autophagy was explored in depth. The lncRNA plasmacytoma variant translocation 1 was found to upregulate Treg autophagy through miR-146a/tumor necrosis factor receptor-associated factor 6 to enhance its immunosuppressive function, thereby alleviating rejection[66]. In addition, in GVHD after HCT, autophagy was found to be essential for the survival of tyrosine-based inhibitory motif domain + Tregs in the bone marrow, and a therapeutic dose of granulocyte-colony-stimulating factor mobilized Tregs into the periphery and induced autophagy to promote reconstruction after stem cell transplantation, which was required for the alleviation of GVHD. Knockout of the key autophagy gene ATG7 in Treg resulted in reduced Treg reconstitution, aggravated GVHD and reduced the survival rate after HCT[67].

MDSCs can be divided into granulocytic MDSCs (G-MDSCs and CD11b + Ly6G + Ly6C-low) and monocytic MDSCs [monocytic-MDSCs (M-MDSCs) and CD11b + Ly6G-Ly6C-high][62]. From the characterization of MDSCs, it is clear that they are studied mainly in tumors. In recent years, the role of MDSCs and their autophagy-related mechanism in transplantation has also attracted attention. A mouse heart transplantation study revealed that mTOR deficiency promotes M-MDSC differentiation and enhances intracellular autophagy, leading to increased immunosuppressive M-MDSC function. The infusion of MTOR-deficient M-MDSCs can prolong cardiac allograft survival and establish immune tolerance in recipient mice by inhibiting T cell activation and inducing Tregs[68].

Effects of autophagy on innate immune cells

Other innate immune cells, such as monocytes/macrophages, neutrophils and natural killer cells, also participate in immune rejection through their own mechanisms. The innate immune system recognizes foreign grafts through pattern recognition receptors, which recognize pathogen-associated molecular patterns and damage-associated molecular patterns[69]. For organ transplantation, cell surface components in the graft, such as polysaccharides and proteins, may be regarded as nonself components by innate immune cells, thereby triggering immune responses. Innate immune cells regulate the activation and differentiation of adaptive immune cells by secreting signaling molecules such as cytokines and chemokines. In addition, macrophages can phagocytose and process antigens in the graft, thereby further activating the adaptive immune response[70]. In addition, innate immune cells are capable of mediating inflammatory responses that lead to local tissue damage and dysfunction at the transplant site. This inflammatory reaction is an important part of graft rejection and one of the major causes of graft failure[71,72].

The role of autophagy in the regulation of innate immune cells cannot be ignored. A study of corneal transplantation in rats revealed that kaempferol could inhibit the activation of the nod-like receptor family pyrin domain containing 3 inflammasome by inducing autophagy, thereby inhibiting macrophage polarization and ultimately reducing corneal graft rejection[73]. In particular, a recent mouse heart transplantation study published in Science showed that monocytes/macrophages can mediate immune memory to alloantigens, which is dependent mainly on cell surface paired Ig-like receptors, and that blocking the binding of paired Ig-like receptors-A to donor MHC-I molecules blocked memory formation and attenuated rejection in kidney and heart grafts[74]. Given the important role of autophagy in immune cell memory formation, it is reasonable to speculate that autophagy is more or less involved in the above studies, which, of course, requires further investigation.

CLINICAL APPLICATION OF TARGETING AUTOPHAGY IN SOLID ORGAN TRANSPLANTATION

With the in-depth understanding of the mechanism of autophagy in immune rejection, there has been a growing interest in exploring its potential clinical applications in organ transplantation. Rapamycin, a classical autophagy inducer, has been widely used as an important immunosuppressant in organ transplantation, which can promote autophagy by inhibiting mTOR[75-77]. Rapamycin has previously been shown to inhibit mTOR by binding to its intracellular receptor FK506 binding protein 12, which inhibits the activation of cyclin D1/CDK4 signaling and thus arrests cell cycle from G1 to S phase and ultimately reducing post-transplant rejection[75,78]. Recent studies indicate that rapamycin may exert immunomodulatory effects by regulating autophagy[73,79,80], although the weight of autophagy in this process remains to be fully elucidated. Other drugs with autophagy-modulating properties have also garnered attention. Hydroxychloroquine (HCQ) is a traditional antimalarial drug and an autophagy inhibitor that blocks autophagosome-lysosome fusion[81,82]. HCQ can interfere with the degradation function of APC lysosomes and block the formation of peptide-MHC protein complexes to inhibit the activation of T cells, which is also considered to be significantly related to autophagy[83]. HCQ has been widely used in the treatment of autoimmune diseases such as systemic lupus erythematosus and scleroderma[84], and has gradually been preliminarily explored in the application of transplantation, especially stem cell transplantation. Two early phase II clinical trials found that HCQ was well tolerated and effective in the prevention and treatment of GVHD after allogeneic bone marrow transplantation[85,86]. However, a subsequent phase III clinical trial confirmed that HCQ had no significant effect on acute/chronic GVHD prevention and survival[87]. A recent retrospective propensity-matched study in kidney transplantation also found that HCQ combined with standard immunosuppressive therapy failed to prevent rejection among renal transplant recipients[88]. 3-MA is another widely used autophagy inhibitor that blocks autophagosome formation by inhibiting the phosphatidylinositide-3kinase-III complex[89-91]. However, due to its high toxicity, poor specificity, and solubility issues, its use is currently limited to experimental research. Additionally, some surgical techniques can modulate autophagy, with a few reported in organ transplantation. An early study on liver transplantation found that ischemic preconditioning can promote the expression of autophagy-related proteins and reduce the incidence of acute and chronic rejection in fatty liver grafts[92]. Overall, there’s a lack of clinical or preclinical studies specifically targeting autophagy in transplant immunology. Drugs like rapamycin which are already used in transplantation, possess multiple signal modulation capabilities[93,94], making it challenging to determine the specific role of autophagy in its immunomodulatory effects.

CONCLUSION

As an important intracellular biological process, autophagy plays an important role in organ transplantation rejection. By regulating the activity and function of immune cells, affecting the production and secretion of cytokines and chemokines, and reducing oxidative stress and the inflammatory response in the graft, the regulation of autophagy can induce immune tolerance, reduce graft rejection, and promote graft survival. However, because the effects of autophagy on different immune cell populations are not consistent, the mechanism of autophagy in organ transplantation is still not fully understood and is controversial, and the regulatory strategy of autophagy still needs to be further optimized and improved. Future research can explore the specific mechanism of autophagy in organ transplantation, develop more specific and effective autophagy regulation strategies, explore the therapeutic value of autophagy in transplantation and rejection after transplantation, and guide individualized immunotherapy to provide new ideas and methods for the clinical practice of organ transplantation.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Transplantation

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade C, Grade C, Grade C

Novelty: Grade B, Grade B, Grade C

Creativity or Innovation: Grade B, Grade B, Grade C

Scientific Significance: Grade B, Grade B, Grade B

P-Reviewer: Amante MF; Ma SH; Majeed HM S-Editor: Bai Y L-Editor: A P-Editor: Zhao YQ

References
1.  Suthanthiran M, Strom TB. Renal transplantation. N Engl J Med. 1994;331:365-376.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 353]  [Cited by in RCA: 334]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
2.  Barker CF, Markmann JF. Historical overview of transplantation. Cold Spring Harb Perspect Med. 2013;3:a014977.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in RCA: 98]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
3.  Bezinover D, Saner F. Organ transplantation in the modern era. BMC Anesthesiol. 2019;19:32.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in RCA: 31]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
4.  Cozzi E, Colpo A, De Silvestro G. The mechanisms of rejection in solid organ transplantation. Transfus Apher Sci. 2017;56:498-505.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in RCA: 25]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
5.  Choudhary NS, Saigal S, Bansal RK, Saraf N, Gautam D, Soin AS. Acute and Chronic Rejection After Liver Transplantation: What A Clinician Needs to Know. J Clin Exp Hepatol. 2017;7:358-366.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in RCA: 102]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
6.  Shibutani ST, Saitoh T, Nowag H, Münz C, Yoshimori T. Autophagy and autophagy-related proteins in the immune system. Nat Immunol. 2015;16:1014-1024.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 348]  [Cited by in RCA: 366]  [Article Influence: 36.6]  [Reference Citation Analysis (0)]
7.  Virgin HW, Levine B. Autophagy genes in immunity. Nat Immunol. 2009;10:461-470.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 328]  [Cited by in RCA: 345]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
8.  Clarke AJ, Simon AK. Autophagy in the renewal, differentiation and homeostasis of immune cells. Nat Rev Immunol. 2019;19:170-183.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 168]  [Cited by in RCA: 251]  [Article Influence: 41.8]  [Reference Citation Analysis (0)]
9.  Glick D, Barth S, Macleod KF. Autophagy: cellular and molecular mechanisms. J Pathol. 2010;221:3-12.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1904]  [Cited by in RCA: 2710]  [Article Influence: 180.7]  [Reference Citation Analysis (0)]
10.  Kim KH, Lee MS. Autophagy--a key player in cellular and body metabolism. Nat Rev Endocrinol. 2014;10:322-337.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 481]  [Cited by in RCA: 706]  [Article Influence: 64.2]  [Reference Citation Analysis (0)]
11.  Yamamoto H, Zhang S, Mizushima N. Autophagy genes in biology and disease. Nat Rev Genet. 2023;24:382-400.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 263]  [Reference Citation Analysis (0)]
12.  Vargas JNS, Hamasaki M, Kawabata T, Youle RJ, Yoshimori T. The mechanisms and roles of selective autophagy in mammals. Nat Rev Mol Cell Biol. 2023;24:167-185.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in RCA: 431]  [Article Influence: 215.5]  [Reference Citation Analysis (0)]
13.  Li X, He S, Ma B. Autophagy and autophagy-related proteins in cancer. Mol Cancer. 2020;19:12.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 304]  [Cited by in RCA: 1022]  [Article Influence: 204.4]  [Reference Citation Analysis (0)]
14.  Cen X, Li Z, Chen X. Ubiquitination in the regulation of autophagy. Acta Biochim Biophys Sin (Shanghai). 2023;55:1348-1357.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
15.  Cao W, Li J, Yang K, Cao D. An overview of autophagy: Mechanism, regulation and research progress. Bull Cancer. 2021;108:304-322.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in RCA: 232]  [Article Influence: 58.0]  [Reference Citation Analysis (0)]
16.  Wood KJ, Goto R. Mechanisms of rejection: current perspectives. Transplantation. 2012;93:1-10.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 248]  [Cited by in RCA: 261]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
17.  Rose AG. Understanding the pathogenesis and the pathology of hyperacute cardiac rejection. Cardiovasc Pathol. 2002;11:171-176.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in RCA: 14]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
18.  Cooper JE. Evaluation and Treatment of Acute Rejection in Kidney Allografts. Clin J Am Soc Nephrol. 2020;15:430-438.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in RCA: 62]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
19.  Rodrigo E, Chedid MF, Segundo DS, Millán JCRS, López-Hoyos M. Acute Rejection Following Kidney Transplantation: State-of-the-Art and Future Perspectives. Curr Pharm Des. 2020;26:3468-3496.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
20.  Becker LE, Morath C, Suesal C. Immune mechanisms of acute and chronic rejection. Clin Biochem. 2016;49:320-323.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in RCA: 26]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
21.  Lai X, Zheng X, Mathew JM, Gallon L, Leventhal JR, Zhang ZJ. Tackling Chronic Kidney Transplant Rejection: Challenges and Promises. Front Immunol. 2021;12:661643.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in RCA: 54]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
22.  Boardman DA, Jacob J, Smyth LA, Lombardi G, Lechler RI. What Is Direct Allorecognition? Curr Transplant Rep. 2016;3:275-283.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in RCA: 18]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
23.  Gökmen MR, Lombardi G, Lechler RI. The importance of the indirect pathway of allorecognition in clinical transplantation. Curr Opin Immunol. 2008;20:568-574.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in RCA: 63]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
24.  Caballero A, Fernandez N, Lavado R, Bravo MJ, Miranda JM, Alonso A. Tolerogenic response: allorecognition pathways. Transpl Immunol. 2006;17:3-6.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in RCA: 20]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
25.  Duneton C, Winterberg PD, Ford ML. Activation and regulation of alloreactive T cell immunity in solid organ transplantation. Nat Rev Nephrol. 2022;18:663-676.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in RCA: 39]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
26.  Ochando J, Fayad ZA, Madsen JC, Netea MG, Mulder WJM. Trained immunity in organ transplantation. Am J Transplant. 2020;20:10-18.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in RCA: 57]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
27.  Madsen JC. Advances in the immunology of heart transplantation. J Heart Lung Transplant. 2017;36:1299-1305.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in RCA: 9]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
28.  Stolp J, Zaitsu M, Wood KJ. Immune Tolerance and Rejection in Organ Transplantation. Methods Mol Biol. 2019;1899:159-180.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in RCA: 20]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
29.  Ghislat G, Lawrence T. Autophagy in dendritic cells. Cell Mol Immunol. 2018;15:944-952.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in RCA: 100]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
30.  Bronietzki AW, Schuster M, Schmitz I. Autophagy in T-cell development, activation and differentiation. Immunol Cell Biol. 2015;93:25-34.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in RCA: 92]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
31.  Sandoval H, Kodali S, Wang J. Regulation of B cell fate, survival, and function by mitochondria and autophagy. Mitochondrion. 2018;41:58-65.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in RCA: 60]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
32.  Gerada C, Ryan KM. Autophagy, the innate immune response and cancer. Mol Oncol. 2020;14:1913-1929.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in RCA: 64]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
33.  Mortensen M, Soilleux EJ, Djordjevic G, Tripp R, Lutteropp M, Sadighi-Akha E, Stranks AJ, Glanville J, Knight S, Jacobsen SE, Kranc KR, Simon AK. The autophagy protein Atg7 is essential for hematopoietic stem cell maintenance. J Exp Med. 2011;208:455-467.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 464]  [Cited by in RCA: 483]  [Article Influence: 34.5]  [Reference Citation Analysis (0)]
34.  Jia W, He MX, McLeod IX, Guo J, Ji D, He YW. Autophagy regulates T lymphocyte proliferation through selective degradation of the cell-cycle inhibitor CDKN1B/p27Kip1. Autophagy. 2015;11:2335-2345.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in RCA: 77]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
35.  Glatigny M, Moriceau S, Rivagorda M, Ramos-Brossier M, Nascimbeni AC, Lante F, Shanley MR, Boudarene N, Rousseaud A, Friedman AK, Settembre C, Kuperwasser N, Friedlander G, Buisson A, Morel E, Codogno P, Oury F. Autophagy Is Required for Memory Formation and Reverses Age-Related Memory Decline. Curr Biol. 2019;29:435-448.e8.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in RCA: 146]  [Article Influence: 24.3]  [Reference Citation Analysis (0)]
36.  Xu X, Araki K, Li S, Han JH, Ye L, Tan WG, Konieczny BT, Bruinsma MW, Martinez J, Pearce EL, Green DR, Jones DP, Virgin HW, Ahmed R. Autophagy is essential for effector CD8(+) T cell survival and memory formation. Nat Immunol. 2014;15:1152-1161.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 358]  [Cited by in RCA: 348]  [Article Influence: 31.6]  [Reference Citation Analysis (0)]
37.  Verghese DA, Yadav A, Bizargity P, Murphy B, Heeger PS, Schröppel B. Costimulatory blockade-induced allograft survival requires Beclin1. Am J Transplant. 2014;14:545-553.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in RCA: 14]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
38.  Chen X, Wang L, Deng Y, Li X, Li G, Zhou J, Cheng D, Yang Y, Yang Q, Chen G, Wang G. Inhibition of Autophagy Prolongs Recipient Survival Through Promoting CD8(+) T Cell Apoptosis in a Rat Liver Transplantation Model. Front Immunol. 2019;10:1356.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in RCA: 18]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
39.  Wang X, Zhu W, Chen H, Li X, Zheng W, Zhang Y, Fan N, Chen X, Wang G. JNK signaling mediates acute rejection via activating autophagy of CD8(+) T cells after liver transplantation in rats. Front Immunol. 2024;15:1359859.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (1)]
40.  Cui J, Yu J, Xu H, Zou Y, Zhang H, Chen S, Le S, Zhao J, Jiang L, Xia J, Wu J. Autophagy-lysosome inhibitor chloroquine prevents CTLA-4 degradation of T cells and attenuates acute rejection in murine skin and heart transplantation. Theranostics. 2020;10:8051-8060.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in RCA: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
41.  Oravecz-Wilson K, Rossi C, Zajac C, Sun Y, Li L, Decoville T, Fujiwara H, Kim S, Peltier D, Reddy P. ATG5-Dependent Autophagy Uncouples T-cell Proliferative and Effector Functions and Separates Graft-versus-Host Disease from Graft-versus-Leukemia. Cancer Res. 2021;81:1063-1075.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
42.  Chong AS. B cells as antigen-presenting cells in transplantation rejection and tolerance. Cell Immunol. 2020;349:104061.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in RCA: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
43.  Montgomery RA, Loupy A, Segev DL. Antibody-mediated rejection: New approaches in prevention and management. Am J Transplant. 2018;18 Suppl 3:3-17.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in RCA: 100]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
44.  Lee BT, Fiel MI, Schiano TD. Antibody-mediated rejection of the liver allograft: An update and a clinico-pathological perspective. J Hepatol. 2021;75:1203-1216.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in RCA: 36]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
45.  Fribourg M, Ni J, Nina Papavasiliou F, Yue Z, Heeger PS, Leventhal JS. Allospecific Memory B Cell Responses Are Dependent on Autophagy. Am J Transplant. 2018;18:102-112.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in RCA: 13]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
46.  Xu Y, Wang Y, Zhang D, Zhang H, Wang Y, Wang W, Hu X. An autophagy-associated diagnostic signature based on peripheral blood for antibody-mediated rejection in renal transplantation. Transpl Immunol. 2024;84:102021.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
47.  Zhang ZW, Wang M, Hu JJ, Xu G, Zhang Y, Zhang N. Decreased Expression of MicroRNA-107 in B Lymphocytes of Patients with Antibody-Mediated Renal Allograft Rejection. Tohoku J Exp Med. 2018;246:87-96.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 5]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
48.  Valečka J, Almeida CR, Su B, Pierre P, Gatti E. Autophagy and MHC-restricted antigen presentation. Mol Immunol. 2018;99:163-170.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in RCA: 52]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
49.  Van Kaer L, Parekh VV, Postoak JL, Wu L. Role of autophagy in MHC class I-restricted antigen presentation. Mol Immunol. 2019;113:2-5.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in RCA: 43]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
50.  Crotzer VL, Blum JS. Autophagy and its role in MHC-mediated antigen presentation. J Immunol. 2009;182:3335-3341.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 169]  [Cited by in RCA: 189]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
51.  Münz C. Autophagy Beyond Intracellular MHC Class II Antigen Presentation. Trends Immunol. 2016;37:755-763.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in RCA: 102]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
52.  Severa M, Giacomini E, Gafa V, Anastasiadou E, Rizzo F, Corazzari M, Romagnoli A, Trivedi P, Fimia GM, Coccia EM. EBV stimulates TLR- and autophagy-dependent pathways and impairs maturation in plasmacytoid dendritic cells: implications for viral immune escape. Eur J Immunol. 2013;43:147-158.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in RCA: 71]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
53.  Hubbard-Lucey VM, Shono Y, Maurer K, West ML, Singer NV, Ziegler CG, Lezcano C, Motta AC, Schmid K, Levi SM, Murphy GF, Liu C, Winkler JD, Amaravadi RK, Rogler G, Dickinson AM, Holler E, van den Brink MR, Cadwell K. Autophagy gene Atg16L1 prevents lethal T cell alloreactivity mediated by dendritic cells. Immunity. 2014;41:579-591.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in RCA: 82]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
54.  Alissafi T, Banos A, Boon L, Sparwasser T, Ghigo A, Wing K, Vassilopoulos D, Boumpas D, Chavakis T, Cadwell K, Verginis P. Tregs restrain dendritic cell autophagy to ameliorate autoimmunity. J Clin Invest. 2017;127:2789-2804.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in RCA: 91]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
55.  Kim MK, Kim J. Properties of immature and mature dendritic cells: phenotype, morphology, phagocytosis, and migration. RSC Adv. 2019;9:11230-11238.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in RCA: 100]  [Article Influence: 16.7]  [Reference Citation Analysis (0)]
56.  Bracho-Sanchez E, Hassanzadeh A, Brusko MA, Wallet MA, Keselowsky BG. Dendritic Cells Treated with Exogenous Indoleamine 2,3-Dioxygenase Maintain an Immature Phenotype and Suppress Antigen-specific T cell Proliferation. J Immunol Regen Med. 2019;5.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in RCA: 14]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
57.  Wildenberg ME, Koelink PJ, Diederen K, Te Velde AA, Wolfkamp SC, Nuij VJ, Peppelenbosch MP, Nobis M, Sansom OJ, Anderson KI, van der Woude CJ, D'Haens GR, van den Brink GR. The ATG16L1 risk allele associated with Crohn's disease results in a Rac1-dependent defect in dendritic cell migration that is corrected by thiopurines. Mucosal Immunol. 2017;10:352-360.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in RCA: 29]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
58.  Lin JC, Huang WP, Liu CL, Lee JJ, Liu TP, Ko WC, Huang YC, Hsu ML, Wu CH, Chen YJ. Sorafenib induces autophagy in human myeloid dendritic cells and prolongs survival of skin allografts. Transplantation. 2013;95:791-800.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in RCA: 14]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
59.  Nguyen H, Kuril S, Bastian D, Kim J, Zhang M, Vaena SG, Dany M, Dai M, Heinrichs JL, Daenthanasanmak A, Iamsawat S, Schutt S, Fu J, Wu Y, Fairlie DP, Atkinson C, Ogretmen B, Tomlinson S, Yu XZ. Complement C3a and C5a receptors promote GVHD by suppressing mitophagy in recipient dendritic cells. JCI Insight. 2018;3:e121697.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in RCA: 22]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
60.  Gabrilovich DI. Myeloid-Derived Suppressor Cells. Cancer Immunol Res. 2017;5:3-8.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 883]  [Cited by in RCA: 1304]  [Article Influence: 163.0]  [Reference Citation Analysis (0)]
61.  Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021;21:485-498.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 623]  [Cited by in RCA: 1047]  [Article Influence: 261.8]  [Reference Citation Analysis (0)]
62.  Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9:162-174.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 5306]  [Cited by in RCA: 5249]  [Article Influence: 328.1]  [Reference Citation Analysis (0)]
63.  Kato H, Perl A. Blockade of Treg Cell Differentiation and Function by the Interleukin-21-Mechanistic Target of Rapamycin Axis Via Suppression of Autophagy in Patients With Systemic Lupus Erythematosus. Arthritis Rheumatol. 2018;70:427-438.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in RCA: 132]  [Article Influence: 18.9]  [Reference Citation Analysis (0)]
64.  Kabat AM, Harrison OJ, Riffelmacher T, Moghaddam AE, Pearson CF, Laing A, Abeler-Dörner L, Forman SP, Grencis RK, Sattentau Q, Simon AK, Pott J, Maloy KJ. The autophagy gene Atg16l1 differentially regulates Treg and TH2 cells to control intestinal inflammation. Elife. 2016;5:e12444.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in RCA: 148]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
65.  Lu J, Liu Y, Wang W, Li P, Qi F. Knockdown of miR-146a in regulatory T cells suppresses heart transplantation rejection in mice by increasing autophagy. Transpl Immunol. 2021;65:101372.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
66.  Lu J, Wang X, Zhang B, Li P, Du X, Qi F. The lncRNA PVT1 regulates autophagy in regulatory T cells to suppress heart transplant rejection in mice by targeting miR-146a. Cell Immunol. 2021;367:104400.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
67.  Le Texier L, Lineburg KE, Cao B, McDonald-Hyman C, Leveque-El Mouttie L, Nicholls J, Melino M, Nalkurthi BC, Alexander KA, Teal B, Blake SJ, Souza-Fonseca-Guimaraes F, Engwerda CR, Kuns RD, Lane SW, Teng M, Teh C, Gray D, Clouston AD, Nilsson SK, Blazar BR, Hill GR, MacDonald KP. Autophagy-dependent regulatory T cells are critical for the control of graft-versus-host disease. JCI Insight. 2016;1:e86850.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in RCA: 45]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
68.  Li J, Chen J, Zhang M, Zhang C, Wu R, Yang T, Qiu Y, Liu J, Zhu T, Zhang Y, Rong R. The mTOR Deficiency in Monocytic Myeloid-Derived Suppressor Cells Protects Mouse Cardiac Allografts by Inducing Allograft Tolerance. Front Immunol. 2021;12:661338.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
69.  Taguchi T, Mukai K. Innate immunity signalling and membrane trafficking. Curr Opin Cell Biol. 2019;59:1-7.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in RCA: 29]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
70.  Wang R, Lan C, Benlagha K, Camara NOS, Miller H, Kubo M, Heegaard S, Lee P, Yang L, Forsman H, Li X, Zhai Z, Liu C. The interaction of innate immune and adaptive immune system. MedComm (2020). 2024;5:e714.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
71.  Ochando J, Ordikhani F, Boros P, Jordan S. The innate immune response to allotransplants: mechanisms and therapeutic potentials. Cell Mol Immunol. 2019;16:350-356.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in RCA: 64]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
72.  Abou-Daya KI, Oberbarnscheidt MH. Innate allorecognition in transplantation. J Heart Lung Transplant. 2021;40:557-561.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 17]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
73.  Tian H, Lin S, Wu J, Ma M, Yu J, Zeng Y, Liu Q, Chen L, Xu J. Kaempferol alleviates corneal transplantation rejection by inhibiting NLRP3 inflammasome activation and macrophage M1 polarization via promoting autophagy. Exp Eye Res. 2021;208:108627.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in RCA: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
74.  Dai H, Lan P, Zhao D, Abou-Daya K, Liu W, Chen W, Friday AJ, Williams AL, Sun T, Chen J, Chen W, Mortin-Toth S, Danska JS, Wiebe C, Nickerson P, Li T, Mathews LR, Turnquist HR, Nicotra ML, Gingras S, Takayama E, Kubagawa H, Shlomchik MJ, Oberbarnscheidt MH, Li XC, Lakkis FG. PIRs mediate innate myeloid cell memory to nonself MHC molecules. Science. 2020;368:1122-1127.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in RCA: 108]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
75.  Yoo YJ, Kim H, Park SR, Yoon YJ. An overview of rapamycin: from discovery to future perspectives. J Ind Microbiol Biotechnol. 2017;44:537-553.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in RCA: 71]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
76.  Chang YY, Juhász G, Goraksha-Hicks P, Arsham AM, Mallin DR, Muller LK, Neufeld TP. Nutrient-dependent regulation of autophagy through the target of rapamycin pathway. Biochem Soc Trans. 2009;37:232-236.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in RCA: 141]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
77.  Li J, Kim SG, Blenis J. Rapamycin: one drug, many effects. Cell Metab. 2014;19:373-379.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 743]  [Cited by in RCA: 904]  [Article Influence: 82.2]  [Reference Citation Analysis (0)]
78.  Waldner M, Fantus D, Solari M, Thomson AW. New perspectives on mTOR inhibitors (rapamycin, rapalogs and TORKinibs) in transplantation. Br J Clin Pharmacol. 2016;82:1158-1170.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in RCA: 66]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
79.  Xiong A, Duan L, Chen J, Fan Z, Zheng F, Tan Z, Gong F, Fang M. Flt3L combined with rapamycin promotes cardiac allograft tolerance by inducing regulatory dendritic cells and allograft autophagy in mice. PLoS One. 2012;7:e46230.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in RCA: 25]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
80.  Amarnath S, Flomerfelt FA, Costanzo CM, Foley JE, Mariotti J, Konecki DM, Gangopadhyay A, Eckhaus M, Wong S, Levine BL, June CH, Fowler DH. Rapamycin generates anti-apoptotic human Th1/Tc1 cells via autophagy for induction of xenogeneic GVHD. Autophagy. 2010;6:523-541.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in RCA: 29]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
81.  Biswas M, Sukasem C. Pharmacogenomics of chloroquine and hydroxychloroquine: current evidence and future implications. Pharmacogenomics. 2023;24:831-840.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
82.  Ferreira PMP, Sousa RWR, Ferreira JRO, Militão GCG, Bezerra DP. Chloroquine and hydroxychloroquine in antitumor therapies based on autophagy-related mechanisms. Pharmacol Res. 2021;168:105582.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in RCA: 84]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
83.  Schrezenmeier E, Dörner T. Mechanisms of action of hydroxychloroquine and chloroquine: implications for rheumatology. Nat Rev Rheumatol. 2020;16:155-166.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 657]  [Cited by in RCA: 869]  [Article Influence: 173.8]  [Reference Citation Analysis (0)]
84.  Nirk EL, Reggiori F, Mauthe M. Hydroxychloroquine in rheumatic autoimmune disorders and beyond. EMBO Mol Med. 2020;12:e12476.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in RCA: 94]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
85.  Khoury H, Trinkaus K, Zhang MJ, Adkins D, Brown R, Vij R, Goodnough LT, Ma MK, McLeod HL, Shenoy S, Horowitz M, Dipersio JF. Hydroxychloroquine for the prevention of acute graft-versus-host disease after unrelated donor transplantation. Biol Blood Marrow Transplant. 2003;9:714-721.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in RCA: 21]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
86.  Gilman AL, Chan KW, Mogul A, Morris C, Goldman FD, Boyer M, Cirenza E, Mazumder A, Gehan E, Cahill R, Frankel S, Schultz K. Hydroxychloroquine for the treatment of chronic graft-versus-host disease. Biol Blood Marrow Transplant. 2000;6:327-334.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in RCA: 68]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
87.  Fong T, Trinkaus K, Adkins D, Vij R, Devine SM, Tomasson M, Goodnough LT, Lopez S, Graubert T, Shenoy S, Dipersio JF, Khoury HJ. A randomized double-blind trial of hydroxychloroquine for the prevention of chronic graft-versus-host disease after allogeneic peripheral blood stem cell transplantation. Biol Blood Marrow Transplant. 2007;13:1201-1206.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in RCA: 17]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
88.  Mohamadou I, Savoye E, Cohen F, Couchoud C, Galichon P. Effect of Hydroxychloroquine Treatment on Kidney Allograft Rejection and Graft Failure. Transplant Proc. 2024;56:1251-1258.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
89.  Wu YT, Tan HL, Shui G, Bauvy C, Huang Q, Wenk MR, Ong CN, Codogno P, Shen HM. Dual role of 3-methyladenine in modulation of autophagy via different temporal patterns of inhibition on class I and III phosphoinositide 3-kinase. J Biol Chem. 2010;285:10850-10861.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 800]  [Cited by in RCA: 914]  [Article Influence: 60.9]  [Reference Citation Analysis (0)]
90.  Seglen PO, Gordon PB. 3-Methyladenine: specific inhibitor of autophagic/lysosomal protein degradation in isolated rat hepatocytes. Proc Natl Acad Sci U S A. 1982;79:1889-1892.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1020]  [Cited by in RCA: 1111]  [Article Influence: 25.8]  [Reference Citation Analysis (0)]
91.  Wu Y, Wang X, Guo H, Zhang B, Zhang XB, Shi ZJ, Yu L. Synthesis and screening of 3-MA derivatives for autophagy inhibitors. Autophagy. 2013;9:595-603.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in RCA: 108]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
92.  Degli Esposti D, Sebagh M, Pham P, Reffas M, Poüs C, Brenner C, Azoulay D, Lemoine A. Ischemic preconditioning induces autophagy and limits necrosis in human recipients of fatty liver grafts, decreasing the incidence of rejection episodes. Cell Death Dis. 2011;2:e111.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in RCA: 62]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
93.  Panwar V, Singh A, Bhatt M, Tonk RK, Azizov S, Raza AS, Sengupta S, Kumar D, Garg M. Multifaceted role of mTOR (mammalian target of rapamycin) signaling pathway in human health and disease. Signal Transduct Target Ther. 2023;8:375.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in RCA: 245]  [Article Influence: 122.5]  [Reference Citation Analysis (0)]
94.  Lamming DW. Inhibition of the Mechanistic Target of Rapamycin (mTOR)-Rapamycin and Beyond. Cold Spring Harb Perspect Med. 2016;6:a025924.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in RCA: 123]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]