Skip to main content
International Journal of Clinical and Experimental Pathology logoLink to International Journal of Clinical and Experimental Pathology
. 2010 Jan 1;3(1):24–38.

HMGB1, an innate alarmin, in the pathogenesis of type 1 diabetes

Shu Zhang 1,*, Jixin Zhong 1,2,*, Ping Yang 1,2, Feili Gong 3, Cong-Yi Wang 2,4
PMCID: PMC2776260  PMID: 19918326

Abstract

HMGB1, an evolutionarily conserved chromosomal protein, was recently re-discovered to act as a “danger signal” (alarmin) to alert the innate immune system for the initiation of host defense or tissue repair. Extracellular HMGB1 can be either passively released from damaged/necrotic cells or secreted by activated immune cells. Upon stimulation, dendritic cells (DCs), macrophages and natural killer (NK) cells secrete high levels of HMGB1 into the intercellular milieu. HMGB1 is potent to target DCs, macrophages, neutrophils and CD4+ T cells. It also upregulates the expression of BCL-XL by which it may prevent the elimination of activated immune cells. As a result, HMGB1 has been suggested to be implicated in the pathogenesis of autoimmune disorders such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and experimental allergic encephalomyelitis (EAE). Given the similarities of autoimmune response against beta cell self-antigens in type 1 diabetes (T1D), in this view we will discuss the possible implications of HMGB1 in T1D pathogenesis. Specifically, we will summarize and update the advancement of HMGB1 in the pathogenesis of autoimmune initiation and progression duringT1D development, as well as islet allograft rejection of diabetic patients after islet transplantation. Elucidation of the role for HMGB1 in T1D pathogenesis would not only enhance the understanding of disease etiology, but also have the potential to shed new insight into the development of therapeutic strategies for prevention or intervention of this disorder.

Keywords: HMGB1, innate alarmin, pathogenesis type 1 diabetes, review

Introduction

Type 1 diabetes (T1D) is an autoimmune disorder characterized by the specific destruction of the insulin secreting beta cells of the pancreatic islets by a certain population of autoreactive immune cells [14]. The early stages of the disease process leading to the development of diabetes are characterized by insulitis, the infiltration of the pancreatic islets by mononuclear cells such as dendritic cells (DCs), macrophages and T cells [5]. The autoimmune etiology of T1D is also manifested by the presence of circulating autoantibodies, specific for beta cell proteins including insulin, glutamic acid decarboxylase 65 (GAD65), and protein tyrosine phosphatase-like protein IA2 (IA-2). These autoantibodies are present in 85–90% of subjects with diabetes at the time of diagnosis [6, 7]. Although it is unclear whether they participate directly in beta cell destruction, they can be served as markers to monitor disease progression [8, 9]. Since autoimmune response progresses many years before the onset of clinical diabetes, studies in T1D pathogenesis for the past several decades have been mainly focused on the role of adaptive immunity. T cells are believed to be the major effector cells responsible for beta cell destruction. CD8+ T cells have been found to be critical for disease pathogenesis in both T1D patients and NOD mice, a mouse model for spontaneous autoimmune diabetes [10, 11]. Autoreactive T cells target beta cell specific self-antigens including insulin and glucose 6-phosphatase catalytic subunit-related protein and consequently mediate beta cell destruction [12, 13]. In addition, CD4+ T cells activated against self-antigens can also promote B cell to produce autoantibodies against those self-antigens. Therefore, T cell mediated adaptive immunity was emphasized so much that the role of innate immunity in disease etiology was overshadowed and ignored.

During evolution from unicellular to multicellular organisms, the immune system has developed a set of mechanisms to sense either non-self component invaded into hosts or damaged self tissues, so that organisms can initiate a defensive response and repair damaged tissue to restore tissue homeostasis, which are also called “inflammatory response”. The inflammatory response is begun with the recognition of those life-threatening events called “danger signals”, which then alert the innate immune system and trigger defensive immune responses. Those “danger signals” can be categorized into pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). PAMPs are defined as exogenous molecules that can alert innate immune system of life-threatening pathogens, such as lipopolysacharide (LPS) and viral RNA. The receptors on immune cells that can recognize PAMPs is so-called pattern recognition receptors (PRRs). A well-known family of PRRs is the toll-like receptor (TLR) family in which each member recognizes a specific set of PAMPs [1416]. On ligation with PAMPs, PRRs transduce activation signals that lead to the production of proinflammatory molecules such as tumour necrosis factor (TNF) to enhance immune response [17, 18]. DAMPs are endogenous molecules now called as alarmins, and once they have released from the damaged cells, they can initiate and promote host inflammatory response and tissue repair [19].

Along with the realization of PAMPs (innate alarmins) that can be recognized by the PPRs expressed on immune cells, there is now compelling evidence suggesting the implication of innate immunity in T1D pathogenesis. Cells infiltrated into insulitis lesion derive from both innate and adaptive immune system such as T lymphocytes, B lymphocytes, DCs, macrophages and natural killer (NK) cells [2023]. Abnormalities in some of the innate cells have been addressed both in animal model and human T1D patients as well as those subjects at risk to the disease [24]. The observation that LPS, viral infections, or generalized activation of APCs delays or prevents the establishment of peripheral tolerance further underscored the importance of innate immunity in the development of autoimmune diabetes [2527]. In this review, we will focus on high-mobility group box 1 (HMGB1), a recently identified endogenous alarmin, in the pathogenesis of autoimmune initiation and progression during T1D development, as well as islet allograft rejection in diabetic patients after islet transplantation. Understanding of these advancements could shed new insight into the possibilities for developing novel therapeutic strategies to mitigate or prevent type 1 diabetes.

The characterization and rediscovery of proinflammatory properties for HMGB1

HMGB1 is a member of high mobility group (HMG) nuclear proteins. This family of non-histone, chromatin associated nuclear proteins was discovered as specific regulators of gene expression more than 35 years ago [28]. HMG proteins are constitutively expressed in the nucleus of eukaryotic cells. They were confirmed to be involved in DNA organization and regulation of transcription. They share functional motifs that bind specific DNA structures and induce conformational changes without specificity for target sequences. They have such structural characteristics as transcripts with long AT-rich 3' untranslated regions and highly negatively charged carboxy-terminals [29].

HMGB1 is probably originated more than 500 million years ago before the split between the animal and plant kingdoms (Figure 1A). It is among the most evolutionarily conserved proteins in the eukaryotic kingdom and shares 100% amino acid (AA) identity between mice and rats, and 99% AA identity between rodents and humans (Figure 1B). HMGB1 has a concentration about 106 molecules per cell and is constitutively expressed in quiescent cells, and a large “pool” of performed HMGB1 is stored in the nucleus [29]. As a nuclear protein, HMGB1 is implicated in diverse cellular functions, including the regulation of nucleosomal structure and stability, and transcription factors binding to their cognate DNA sequences [2934]. The binding activity of HMGB1 to DNA is regulated by the two 80-amino acid DNA binding domains, the A-box and B-box, with each structurally represented as three α-helices in a characteristic L-shaped fold [35] (Figure 2). In addition to A- and B-box, there is an acidic tail in the C-terminal of HMGB1. The C-terminal acidic tail is important for the transcription stimulatory function of HMGB1 [3538]. The two boxes bind to the minor groove of chromatin thus modifying the DNA architecture. This facilitates the binding of regulatory proteins of various transcription factors to their cognate sequences, including the steroid/nuclear hormones progesterone [39] and estrogen [40, 41], HOX proteins [42], p53, homeobox-containing proteins, recombination activating gene 1/2 (RAG1/2) proteins and transcription factor II B [43]. Mice deficient for HMGB1 are viable for several days if given glucose parenterally, then waste away probably due to the inactivation of glucocorticoid receptor (GR) transcribed genes [44]. Phenotypic features include small size, ruffled and disorganized fur, long hind paws, and absence of fat. Cell lines lacking HMGB1 grow normally, but the activation of gene expression by different factors including glucocorticoid receptor, is impaired [44].

Figure 1.

Figure 1

Sequence analysis showing evolutionary conservation of HMGB1 between diverse species. A: A phylogenetic tree showing evolutionary relationships of the HMGB1 gene between different species. The phylogenetic tree was constructed according to the calculation of the best match for the selected sequences. B: HMGB1 amino acid sequence alignment showing evolutionary conservation between diverse species. Sequence homology: black, 100% identical; pink, >75% identical; blue, >50% identical; and white, 0% identical.

Figure 2.

Figure 2

Functional domains within the HMGB1 amino acid sequence. The full-length HMGB1 contains 2 homogenous domains (A- and B-box) and an acidic C-terminal tail. The B-box is associated with its properties relevant to proinflammatory activity and RAGE binding, while the A-box is a specific antagonist by which it inhibits the proinflammatory properties of HMGB1. The C-terminal acidic tail is required for transcription stimulatory function of HMGB1.

In late 1990s, in the search for a broader therapeutic window for the treatment of sepsis and endotoxemia, Wang and colleagues re-discovered HMGB1 as a late mediator of endotoxin lethality in a murine model [45], and blockade of HMGB1 by the administration of neutralizing antibodies protects against LPS lethality in mice. Follow-up studies demonstrated that extracellular HGMB1 acts as a potent innate “danger signal” to alert the innate immune system for the exogenous invasive microorganisms, the endogenous tissue injury or the presence of intercellular inflammatory mediators, which then initiates host defense or tissue repair. B-box domain is important for the proinflammatory properties of HMGB1 including cytokine release [46, 47]. Instead of possessing proinflammatory properties, the A-box competes with full length HMGB1 for binding sites leading to attenuation of the inflammatory cascade [48] (Figure 2).

Two distinct mechanisms have subsequently been proposed for cells to liberate HMGB1 into the extracellular milieu. The damaged/necrotic cells can passively release HMGB1 into extracellular milieu, which represents an intracel-lular marker selected by the innate immune system to recognize tissue damage and initiate reparative responses [49, 50]. Caspase-dependent oxidization of HMGB1 by reactive oxygen species (ROS) has been suggested to be critical for tolerance induction by damaged or apoptotic cells [51]. The second mechanism is the “active secretion” of HMGB1 by activated immune cells to mimic the necrotic process and activate innate immune response during an immunological challenge. Unlike passive release, HMGB1 active secretion has been shown to be associated with extensive acetylation of lysine residues [52]. Extracellular HMGB1 exerts not only paracrine activity, but also autocrine activity on the cells from which it is secreted to enhance chemotaxis and innate immune response, and subsequently to initiate and promote adaptive immune response [53].

Receptors associated with HMGB1 signaling pathways

There are several important receptors have thus far been characterized to be implicated in HMGB1 signaling, including the receptor for advanced glycation end products (RAGE) and some members of the Toll-like family of receptors. RAGE is thought to be one of the primary receptors for HMGB1 [54]. RAGE is a transmembrane protein expressed at low levels in normal tissues, but it is upregulated at sites where its ligands accumulate [55]. RAGE expression is detected on monocytes, macrophages, neurons, and endothelial cells, as well as on a variety of tumor cells [56, 57]. Binding of RAGE by HMGB1 can activate both CDC42/Rac pathway and MAPKs-NFκB pathway [56, 58, 59]. These two HMGB1 signaling pathways then through RAGE promotes chemotaxis, production of cytokines relevant to NFκB activation [60, 61], activation of endothelial cells [62, 63], maturation and migration of immune cells [47, 6470].

Despite the characterization of RAGE as the receptor for HMGB1, RAGE alone, however, could not explain all the observed effects of HMGB1, suggesting the existence of additional receptor(s) relevant to HMGB1 signaling. Consistent with this assumption, follow up studies further characterized that toll-like receptors 2 (TLR2) and 4 (TLR4) are involved in HMGB1 signaling [7173]. TLR4 is the main receptor for endotoxin, whereas TLR2 responds to Gram-positive components and fungi [74]. HMGB1 signaling through TLR2 and TLR4 is mediated by Rac1/PI3K/CDC42 pathway and MyD88 dependent NFκB activation pathway [61, 7176]. More recently, Ivanov and colleagues further demonstrated that HMGB1 acts as a CpG-ODN-binding protein, by which it interacts and preassociates with TLR9 in the endoplasmic reticulum-Golgi intermediate compartment (ERGIC), and as a result, forms a complex within specialized vesicles [77]. CpG-ODN stimulates macrophages and DCs to secrete high levels of HMGB1, which in turn accelerates the delivery of CpG-ODNs to its receptor, leading to a TLR9-dependent augmentation of IL-6, IL-12, and TNFα secretion. It seems that HMGB1 does not affect the uptake of CpG DNA or its entry into early endosomes, but rather accelerates the redistribution of TLR9 from the endoplasmic reticulum to the early endosomes in response to CpG DNA [77].

HMGB1 in the pathogenesis of autoimmune initiation during T1D development

The initiation of autoimmune response during T1D development is a multifactorial and complex process that requires genetic predisposition to synergize with unknown exogenous and/or endogenous triggers [7883]. Once triggered by those factors, macrophages and DCs infiltrated into islets and followed shortly thereafter by the recruitment of T and B lymphocytes [20–23, 84, 85]. Apoptosis is a normal physiological process contributing to tissue turnover during the development and regulation of tissue homeostasis in the adult [86]. Unlike necrosis, apoptosis is usually considered to be a non-inflammatory process. However, recent investigations ranging from animal models to human pathology lend support to the view that apoptosis plays a pivotal role in the development of autoimmunity [8793]. Apoptotic cells are a source of autoantigens, and apoptosis has been suggested in certain conditions, contributing to autoimmune response in multiple autoimmune disorders including systemic lupus erythematosus (SLE) [89, 91, 94], rheumatoid arthritis (RA) [95, 96], and experimental allergic encephalomyelitis (EAE) [97, 98]. A number of recent studies indicate that HMGB1 can be passively released from late apoptotic cells in a cell type dependent manner [99, 100], probably through a mechanism by binding to some nuclear molecules such as DNA that can be leaked out from apoptotic cells [99, 101, 102].

To address whether HMGB1 can be passively released from late apoptotic beta cells, we first induced NIT-1 cell (a NOD-derived β cell line) apoptosis by combination of cytokines (100U/ml INFY, 10U/ml IL-1β and 100U/ml TNFα) [103]. After 72h of treatment, around 73% of NIT-1 cells became apoptotic, and 79% of which were undergoing secondary necrosis as determined by propidium iodide (PI) and annexin V staining. Western blot analysis of culture supernatants derived from cells treated with cytokines detected high levels of HMGB1, while HMGB1 was absent from control culture supernatants, indicating passive release of HMGB1 from late apoptotic beta cells [103]. To further address the question, we performed follow-up studies to examine the effect of late apoptotic beta cells on NOD DC activation and functionality. NIT-1 cells were first induced apoptosis by UV treatment and the resulting proapoptotic cells were subsequently labeled with carboxyfluorescein succinimidyl ester (CFSE). Immature NOD bone marrow-derived DCs (BMDCs) were then co-cultured with CSFE-labeled apoptotic NIT-1 cells at 1:20 ratio for 4h, followed by staining DCs with a PE-labeled anti-CD11c antibody. Flow cytometry analysis indicated that approximately 81% of DCs were double positive for CD11c and CFSE, indicating that NOD DCs have high potency for uptake of apoptotic NIT-1 cells (unpublished data). Confocal microscopic analysis of DCs in different layers further confirmed that some DCs can actually ingest more than one apoptotic NIT-1 cells. More interestingly, NOD DCs after internalizing CSFE-labeled NIT-1 cells show a matured phenotype as manifested by high levels of surface MHC II expressions (unpublished data). Together, all of these observations provide an overall support for the scenario that HMGB1 originated from late apoptotic beta cells has high potency to stimulate NOD DC activation, which could then initiate autoimmune response against beta cell self antigens leading to beta cell destruction.

Previous studies including ours have consistently demonstrated that NOD neonates show abnormal beta cell apoptosis during beta mass turnover after birth, probably due to defective clearance of apoptotic cells in NOD mice [104107]. In situ TUNEL assays of pancreatic sections originated from NOD and B6 neonates indicate that NOD newborns have at least 2-fold higher apoptosis as compared to that of B6 newborns (Figure 3 A vs. B). Insulin co-staining further confirmed that these apoptotic cells were actually insulin-producing beta cells (Figure 3A, inset figure, where insulin stained blue, apoptotic cell stained red, and the apoptotic cell was indicated by an arrow). Since excessive apoptosis is usually associated with secondary necrosis [108], in vivo propidium iodide (PI) staining was then carried out to check beta cell necrosis by injection of 15ul PI (20μg per gram of body weight) into 2wk-old NOD and control newborns via tail vein. Pancreatic frozen sections were prepared after 3h injection. In line with previous observations, PI positive cells (necrotic cells) were observed in the sections of NOD neonates, while necrosis was almost undetectable in B6 neonates (Figure 3, C vs. D). Since HMGB1 passive release is associated with secondary necrosis, in situ HMGB1 immunostaining was then conducted in pancreatic sections of NOD neonates. Unlike B6 neonate islets in which HMGB1 is solely localized in the nucleus of islet cells, some islet cells in NOD neonate show condensed nuclei, plasma membrane blebbings, and displayed positive nuclear and cytoplasmic staining of HMGB1 (unpublished data), indicating HMGB1 passive release from secondary necrotic beta cells. These passively released HMGB1 in turn, may function as an innate alarmin to activate DCs for the presentation of beta cell self-antigens, predisposing to the development of autoimmune diabetes. Although the experimental evidence is currently lacking, it would not be difficult to extrapolate that blockade of HMGB1 during NOD neonate beta mass turnover would prevent or delay insulitis, a typical characteristic of autoimmune response during T1D development.

Figure 3.

Figure 3

Altered beta-cell apoptosis during NOD physiological beta mass turnover after birth. A & B: TUNEL assay of pancreatic beta cell apoptosis during NOD and B6 neonate beta mass turnover. Insulin costaining indicates that the apoptotic cells are beta cells (A, the apoptotic cell is indicated by an arrow). C & D: Propidium Iodide (PI) staining of secondary necrotic beta cells in pancreatic sections originated from neonates undergoing beta mass turnover.

HMGB1 in autoimmune progression during T1D development

Upon the presence of activation signals derived from PRR transduction, beta cell-specific antigens would be recognized by APCs which then become activated to initiate autoimmune response. Once autoimmune response initiated, disease undergoes a progression stage in which adaptive immunity is activated by self-reactive APCs and plays a major role in beta cell destruction. The subsequent beta cell destruction would further increase the amount of self-antigens for APCs which then activate additional autoreactive T and B cells. Therefore, as T1D progresses in severity, so does the number of autoreactive T and B cell clones, known as epitope spreading [109]. Once the disease enters the progression stage of autoimmune-mediated beta cell destruction, the difficulty of therapy will be dramatically increased. Unfortunately, at the time of clinical diagnosis all diabetic patients have already been in the progression stage. Therefore, understanding the mechanism of autoimmune process in diabetes progression stage is pivotal for the development of novel therapeutic strategies.

As aforementioned, in addition to passive release from damaged beta cells, HMGB1 can also be secreted by activated immune cells such as DCs [64, 66]. By examining HMGB1 subcellular localization in DCs we have clearly demonstrated that NOD DCs have high potency to actively secrete HMGB1 upon stimulation. Before stimulation, HMGB1 was solely localized in the nucleus, while a major proportion of HMGB1 translocated into the cytoplasm upon LPS or TNFα/IFNγ stimulation. In line with this observation, Western blot analysis of culture supernatants detected high levels of HMGB1 derived from stimulated NOD DCs in a time-dependent manner [103]. To demonstrate the evidence of HMGB1 active secretion by NOD islet infiltrated DCs, in situ HMGB1 immunostaining was carried out using pancreatic sections originated from NOD mice with insulitis (14wk-old). It was found that a large proportion of HMGB1 had translocated into the cytoplasm in the islet infiltrated DCs as they were also positive for CD11c staining, demonstrating HMGB1 active secretion by activated autoreactive DCs, which could then enhance autoimmune progression [103].

Two sets of experiments were carried out to demonstrate the direct evidence for HMGB1 in the pathogenesis of autoimmune progression during T1D development. An endotoxin-free HMGB1 neutralizing antibody was used for the purpose. The study was first carried out in NOD mice at the early stage of autoimmunity (8wk-old). The blocking antibody was administered into those mice once a week (600μg/mouse) for 18 consecutive weeks and then monitored for diabetes onset up to 35wk-old. Remarkably, HMGB1 antibody treatment reduced diabetes incidence by almost 2-fold. Furthermore, the treatment also significantly delayed the onset of diabetes. In average, the age for onset of diabetes in HMGB1 antibody treated mice was 28.7 ± 3.4wk, while the control IgG treated mice was only 18.4 ± 3.1wk [103]. Examination of insulitis severity by histological analysis of pancreatic sections further confirmed that the extent of insulitis in HMGB1 antibody treated mice was significantly less severe than the control IgG treated mice at 12wk, 15wk and 18wk time point examined. To demonstrate the implication of HMGB1 in late stage of autoimmunity, HMGB1 blocking antibody was then administered into 12wk-old NOD mice by then autoimmune infiltration in these mice had already progressed for at least 5wk [110]. Similar as above, the mice were administered with HMGB1 blocking antibody once a week for 13 consecutive weeks and then monitored for diabetes onset. Consistently, blockade of HMGBl significantly reduced diabetes incidence, but unlike animals in the early stage of autoimmunity, the treatment failed to delay the onset of diabetes [103].

Given the critical role of DCs in T1D associated autoimmunity, we first checked the impact of HMGB1 blocking antibody on DCs, the most potent APCs known today. It was found that antibody treatment reduced the number and maturation of a subset of DCs probably associated with auto-antigen presentation to naïve T cells in the pancreatic lymph nodes (PLN). We also noticed a skewed population of CD11c+CD8a+ DCs in mice after blockade of HMGB1. As those cells showed less potency to stimulate alloimmune response, they were probably responsible for inducing peripheral tolerance to tissue associated antigens [111]. In line with this assumption, there was a significant increase for the CD4+Foxp3+ regulatory T cells (Tregs) in the PLN of mice treated with neutralizing antibody [103]. Taken together, these data demonstrated strong evidence indicating that HMGB1 is implicated in the progression of autoimmune response during T1D development.

HMGB1 in islet allograft rejection of diabetic patients after islet transplantation

Current therapeutic strategy for T1D is lifelong commitment of exogenous insulin, monitoring blood glucose, taking healthy foods plus maintaining a healthy weight. However, insulin therapy and healthy lifestyle cannot provide adequate glycemic control for some patients with severe type 1 diabetes, or with complications that cannot be efficiently managed by insulin. Furthermore, insulin administration cannot control blood glucose accurately enough to maintain at a perfect level even with careful insulin administration, and thus would result in chronic complications. It has been shown that even transient hyperglycemia can lead to persistent epigenetic changes predisposing to the development of diabetic complications. In battle against type 1 diabetes, pancreatic transplantation has been considered to offer a successful therapeutic approach for many years [112, 113]. However, similar as other solid organ transplantation, lifelong immunosuppression is required and donor organs are in short supply [79]. An alternative approach, transplantation of donor islets, was later established, which seems to be a promising therapeutic approach to tightly maintain blood glucose levels for patients with type 1 diabetes [114]. Some centers have actually achieved high rate for long-term independence of exogenous insulin. Nevertheless, the total success rate is still not high enough [115, 116].

The major problem hampering islet transplantation is islet allograft dysfunction caused by rejection although it is less severe than solid organ transplantation. Similar as the study field of autoimmunity, the role of adaptive immunity in allograft rejection has been extensively studied for decades. It has been suggested that acute allograft rejection is principally a T cell-mediated adaptive immune response [117]. However, the contribution of innate immunity to allograft rejection has generally been ignored until the discovery of the TLR system and the subsequent realization of innate immunity in driving and shaping adaptive immunity. Early islet loss is considered to be responsible for islet allograft dysfunction, and it is believed to be attributable to inflammatory events in which innate immunity plays an important role [118]. Early loss of islet results in the release of alloantigen and some endogenous alarmins such as HMGB1 which then initiate alloimmune response against islet allograft. Therefore, similar as its role in autoimmunity relevant to T1D pathogenesis, HMGB1 could be also implicated in the initiation and progression of islet allograft rejection.

It has been suggested that ischemia/reperfusion is a major pathophysiological component of acute allograft malfunction after transplantation, which includes direct cellular damage and acute allograft rejection. To demonstrate the implication of HMGB1 in this process, we examined HMGB1 passive release from allograft damaged cells and active secretion by allograft infiltrated immune cells. By analysis of cytoplasmic proteins originated from cardiac allografts that underwent around one hour of cold ischemic insult, we have obtained strong evidence supporting that HMGB1 can be passively released from allograft damaged cells [117]. Consistent with this observation, HMGB1 has been found to be critical in mediating hepatic injury after murine ischemia/reperfusion [119]. We further confirmed that allograft infiltrated immune cells actively secrete high levels of HMGB1 into the intercellular milieu through immuno histochemical analysis of cardiac allograft sections that underwent acute rejection [117]. A steady increase for HMGB1 expression in the allografts after transplantation was noticed by comparative analysis of HMGB1 temporal expression changes between syngeneic and allogeneic cardiac grafts, and the increase of HMGB1 expression in allografts is accompanied by the interstitial infiltration and active secretion of HMGB1 by infiltrated immune cells. In sharp contrast, the increase for HMGB1 expression in syngenic grafts was only noticed after first 3-day of transplantation, and after which, the expression of HMGB1 dropped back to the normal levels. Together, these data indicate that HMGB1 is implicated in acute rejection of allografts and tissue repair of syngeneic grafts. Based on these observations, an endo-toxin-free recombinant peptide encoding HMGB1 A-box (rA-box) was used to treat recipients after cardiac allograft transplantation. Notably, rA-box treatment significantly prolonged allograft survival (12.5 ± 1.87 days vs. 6.5 ± 1.04 days) [117]. In line with this result, recipient mice received rA-box displayed well preserved myocardium and significant reduced inflammatory infiltration after day 7 of transplantation. This protective effect was associated with suppressed production of pathogenic CD8+IFNγ+ (Tc1) and CD4+IFNγ+ (Th1) cells. Similarly, blockade of RAGE, a primary receptor for HMGB1, prevented cardiac allograft rejection [120].

The implication of TLRs in the pathogenesis of allograft rejection further supports a role for HMGB1 in islet allograft rejection. Chen et al. revealed that inhibition of TLR signaling promoted the acceptance of allografts that are resistant to tolerance induction such as skin, whereas administration of TLR agonist during perioperative period abrogated tolerance [121, 122]. MyD88 deficiency inhibited the activation, migration of macrophages in mice transplanted with porcine islet xenografts [123]. Loss of MyD88 signaling reduced mature DCs in the draining lymph nodes and prevented the development of T cell response against graft, and thus induced acceptance of allograft [124, 125]. More interestingly, TLRs on beta cells are also involved in islet allograft rejection. Enhanced TLR expression was observed on porcine islets exposed to macrophages [126]. Goldberg et al. reported that TLR4 expression was elevated on beta cells during islet isolation process, and enhanced TLR4 promoted beta cell death and graft rejection after islet transplantation [124]. In contrast, suppression of TLR4 on donor beta cells by either carbon monoxide (CO) exposure or pre-infection with a TLR4 dominant negative virus prevented the initiation of inflammation and subsequent islet allograft rejection [124]. Similarly, RAGE, another receptor for HMGB1, is also implicated in the rejection of islet allografts. Loss of RAGE or blockade of RAGE by its antagonist TTP488 in recipient mice suppressed T cell response and prolonged islet allograft survival [127]. All together, similar as its role in autoimmune response during T1D development, HMGB1 also plays a pivotal role in the initiation and progression of islet allograft rejection and contributes to islet allograft dysfunction after transplantation.

Conclusion remarks

HMGB1, an evolutionarily conserved chromosomal protein, was recently re-discovered to be an alarmin that alerts the innate immune system to initiate defensive response. Monocytes, macrophages, NK, myeloid and plasmacytoid DCs secrete high levels of HMGB1 in response to pathogen or damage associated molecules. Extracellular HMGB1 targets myeloid DCs, plasmacytoid DCs, macrophages, neutrophils and CD4+ T cells. HMGB1 increases maturation and cytokine secretion from myeloid and plasmacytoid DCs that have been stimulated with TLR ligands [53]. Upon interaction, DCs secrete IL-18, causing NK cells to secrete HMGB1, which in return acts back on DCs to prevent them from NK cell mediated lysis [53] (Figure 4). Based on its importance in orchestrating and modulating adaptive immune responses, here we have discussed the possible implications of HMGB1 in multiple stages of autoimmunity, from innate recognition of self antigens to progression of adaptive autoimmune response against beta cells, as well as islet allograft rejection after transplantation in diabetic patients. The challenge for future studies would be the development of high potent HMGB1 blockade to evaluate its impact on reversal of overt type 1 diabetes and recurrence of autoimmunity after islet transplantation.

Figure 4.

Figure 4

The immuno-regulatory properties for extracellular HMGB1. HMGB1 secretion by activated immune cells is indicated by blue arrows. Extracellular HMGB1 targets myeloid DCs, plasmacytoid DCs, macrophages, neutrophils and CD4+ T cells via the corresponding receptor(s) expressed on their surface. HMGB1 has also been found to up regulate the expression of BCL-XL, which may in turn prevent the elimination of activated autoreactive immune cells.

Acknowledgments

Our research is supported by grants from the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), the Juvenile Diabetes Research Foundation International (JDRFI), the American Diabetes Association (ADA), and the SECTR award from South Carolina and Georgia research program to CYW, and in part supported by the National Key Basic Research Program of China (2007CB512402) to FG. The authors declare that they have no competing financial interest.

References

  • 1.Wang CY, Podolsky R, She JX. Genetic and functional evidence supporting SUMO4 as a type 1 diabetes susceptibility gene. Ann N Y Acad Sci. 2006;1079:257–267. doi: 10.1196/annals.1375.039. [DOI] [PubMed] [Google Scholar]
  • 2.Wang CY, She JX. SUMO4 and its role in type 1 diabetes pathogenesis. Diabetes Metab Res Rev. 2008;24:93–102. doi: 10.1002/dmrr.797. [DOI] [PubMed] [Google Scholar]
  • 3.Li M, Guo D, Isales CM, Eizirik DL, Atkinson M, She JX, Wang CY. SUMO wrestling with type 1 diabetes. J Mol Med. 2005;83:504–513. doi: 10.1007/s00109-005-0645-5. [DOI] [PubMed] [Google Scholar]
  • 4.Wang CY, Han J, She JX. World Scientific, editor. Genetic factors for human type 1 diabetes. Current Topics in Human Genetics (Studies in Complex Diseases) 2008;24:693–727. [Google Scholar]
  • 5.Kelly MA, Rayner ML, Mijovic CH, Barnett AH. Molecular aspects of type 1 diabetes. Mol Pathol. 2003;56:1–10. doi: 10.1136/mp.56.1.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Palmer JP, Asplin CM, Clemons P, Lyen K, Tatpati O, Raghu PK, Paquette TL. Insulin antibodies in insulin-dependent diabetics before insulin treatment. Science. 1983;222:1337–1339. doi: 10.1126/science.6362005. [DOI] [PubMed] [Google Scholar]
  • 7.Leslie RD, Atkinson MA, Notkins AL. Autoantigens IA-2 and GAD in Type I (insulin-dependent) diabetes. Diabetologia. 1999;42:3–14. doi: 10.1007/s001250051105. [DOI] [PubMed] [Google Scholar]
  • 8.Bingley PJ, Bonifacio E, Williams AJ, Genovese S, Bottazzo GF, Gale EA. Prediction of IDDM in the general population: strategies based on combinations of autoantibody markers1. Diabetes. 1997;46:1701–1710. doi: 10.2337/diab.46.11.1701. [DOI] [PubMed] [Google Scholar]
  • 9.LaGasse JM, Brantley MS, Leech NJ, Rowe RE, Monks S, Palmer JP, Nepom GT, McCulloch DK, Hagopian WA. Successful prospective prediction of type 1 diabetes in schoolchildren through multiple defined autoantibodies: an 8-year follow-up of the Washington State Diabetes Prediction Study. Diabetes Care. 2002;25:505–511. doi: 10.2337/diacare.25.3.505. [DOI] [PubMed] [Google Scholar]
  • 10.Bottazzo GF, Dean BM, McNally JM, MacKay EH, Swift PG, Gamble DR. In situ characterization of autoimmune phenomena and expression of HLA molecules in the pancreas in diabetic insulitis. N Engl J Med. 1985;313:353–360. doi: 10.1056/NEJM198508083130604. [DOI] [PubMed] [Google Scholar]
  • 11.Conrad B, Weidmann E, Trucco G, Rudert WA, Behboo R, Ricordi C, Rodriquez-Rilo H, Finegold D, Trucco M. Evidence for superantigen involvement in insulin-dependent diabetes mellitus aetiology. Nature. 1994;371:351–355. doi: 10.1038/371351a0. [DOI] [PubMed] [Google Scholar]
  • 12.Wong FS, Karttunen J, Dumont C, Wen L, Visintin I, Pilip IM, Shastri N, Pamer EG, Janeway CA., Jr Identification of an MHC class I-restricted autoantigen in type 1 diabetes by screening an organ-specific cDNA library. Nat Med. 1999;5:1026–1031. doi: 10.1038/12465. [DOI] [PubMed] [Google Scholar]
  • 13.Lieberman SM, Evans AM, Han B, Takaki T, Vinnitskaya Y, Caldwell JA, Serreze DV, Shabanowitz J, Hunt DF, Nathenson SG, Santamaria P, DiLorenzo TP. Identification of the beta cell antigen targeted by a prevalent population of pathogenic CD8+ T cells in autoimmune diabetes. Proc Natl Acad Sci USA. 2003;100:8384–8388. doi: 10.1073/pnas.0932778100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Akira S, Takeda K, Kaisho T. Toll-like receptors: critical proteins linking innate and acquired immunity. Nat Immunol. 2001;2:675–680. doi: 10.1038/90609. [DOI] [PubMed] [Google Scholar]
  • 15.Lemaitre B, Nicolas E, Michaut L, Reichhart JM, Hoffmann JA. The dorsoventral regulatory gene cassette spatzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell. 1996;86:973–983. doi: 10.1016/s0092-8674(00)80172-5. [DOI] [PubMed] [Google Scholar]
  • 16.Stein D, Roth S, Vogelsang E, Nusslein-Volhard C. The polarity of the dorsoventral axis in the Drosophila embryo is defined by an extracellular signal. Cell. 1991;65:725–735. doi: 10.1016/0092-8674(91)90381-8. [DOI] [PubMed] [Google Scholar]
  • 17.Esen N, Kielian T. Central role for MyD88 in the responses of microglia to pathogen-associated molecular patterns. J Immunol. 2006;176:6802–6811. doi: 10.4049/jimmunol.176.11.6802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hajishengallis G, Sojar H, Genco RJ, DeNardin E. Intracellular signaling and cytokine induction upon interactions of Porphyromonas gingivalis fimbriae with pattern-recognition receptors. Immunol Invest. 2004;33:157–172. doi: 10.1081/imm-120030917. [DOI] [PubMed] [Google Scholar]
  • 19.Bianchi ME. DAMPs, PAMPs and alarmins: all we need to know about danger. J Leukoc Biol. 2007;81:1–5. doi: 10.1189/jlb.0306164. [DOI] [PubMed] [Google Scholar]
  • 20.Pietropaolo M, Barinas-Mitchell E, Kuller LH. The heterogeneity of diabetes: unraveling a dispute: is systemic inflammation related to islet autoimmunity? Diabetes. 2007;56:1189–1197. doi: 10.2337/db06-0880. [DOI] [PubMed] [Google Scholar]
  • 21.Delovitch TL, Singh B. The nonobese diabetic mouse as a model of autoimmune diabetes: immune dysregulation gets the NOD. Immunity. 1997;7:727–738. doi: 10.1016/s1074-7613(00)80392-1. [DOI] [PubMed] [Google Scholar]
  • 22.Katz JD, Benoist C, Mathis D. T helper cell subsets in insulin-dependent diabetes. Science. 1995;268:1185–1188. doi: 10.1126/science.7761837. [DOI] [PubMed] [Google Scholar]
  • 23.Rosmalen JG, Martin T, Dobbs C, Voerman JS, Drexhage HA, Haskins K, Leenen PJ. Subsets of macrophages and dendritic cells in non-obese diabetic mouse pancreatic inflammatory infiltrates: correlation with the development of diabetes. Lab Invest. 2000;80:23–30. doi: 10.1038/labinvest.3780004. [DOI] [PubMed] [Google Scholar]
  • 24.Beyan H, Buckley LR, Yousaf N, Londei M, Leslie RD. A role for innate immunity in type 1 diabetes? Diabetes Metab Res Rev. 2003;19:89–100. doi: 10.1002/dmrr.341. [DOI] [PubMed] [Google Scholar]
  • 25.Ehl S, Hombach J, Aichele P, Rulicke T, Odermatt B, Hengartner H, Zinkernagel R, Pircher H. Viral and bacterial infections interfere with peripheral tolerance induction and activate CD8+ T cells to cause immunopathology. J Exp Med. 1998;187:763–774. doi: 10.1084/jem.187.5.763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Maxwell JR, Campbell JD, Kim CH, Vella AT. CD40 activation boosts T cell immunity in vivo by enhancing T cell clonal expansion and delaying peripheral T cell deletion. J Immunol. 1999;162:2024–2034. [PubMed] [Google Scholar]
  • 27.Vella AT, McCormack JE, Linsley PS, Kappler JW, Marrack P. Lipopolysaccharide interferes with the induction of peripheral T cell death. Immunity. 1995;2:261–270. doi: 10.1016/1074-7613(95)90050-0. [DOI] [PubMed] [Google Scholar]
  • 28.Goodwin GH, Sanders C, Johns EW. A new group of chromatin-associated proteins with a high content of acidic and basic amino acids. Eur J Biochem. 1973;38:14–19. doi: 10.1111/j.1432-1033.1973.tb03026.x. [DOI] [PubMed] [Google Scholar]
  • 29.Bustin M. Regulation of DNA-dependent activities by the functional motifs of the high-mobility-group chromosomal proteins1. Mol Cell Biol. 1999;19:5237–5246. doi: 10.1128/mcb.19.8.5237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Bianchi ME, Beltrame M, Paonessa G. Specific recognition of cruciform DNA by nuclear protein HMG1. Science. 1989;243:1056–1059. doi: 10.1126/science.2922595. [DOI] [PubMed] [Google Scholar]
  • 31.Hill DA, Reeves R. Competition between HMG-I(Y), HMG-1 and histone H1 on four-wayjunction DNA. Nucleic Acids Res. 1997;25:3523–3531. doi: 10.1093/nar/25.17.3523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hill DA, Pedulla ML, Reeves R. Directional binding of HMG-I(Y) on four-wayjunction DNA and the molecular basis for competitive binding with HMG-1 and histone H1. Nucleic Acids Res. 1999;27:2135–2144. doi: 10.1093/nar/27.10.2135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Locker D, Decoville M, Maurizot JC, Bianchi ME, Leng M. Interaction between cisplatin-modified DNA and the HMG boxes of HMG 1: DNase I footprinting and circular dichroism. J Mol Biol. 1995;246:243–247. doi: 10.1006/jmbi.1994.0079. [DOI] [PubMed] [Google Scholar]
  • 34.Stros M, Reich J. Formation of large nucleo-protein complexes upon binding of the high-mobility-group (HMG) box B-domain of HMG1 protein to supercoiled DNA1. Eur J Biochem. 1998;251:427–434. doi: 10.1046/j.1432-1327.1998.2510427.x. [DOI] [PubMed] [Google Scholar]
  • 35.Weir HM, Kraulis PJ, Hill CS, Raine AR, Laue ED, Thomas JO. Structure of the HMG box motif in the B-domain of HMG1. EMBO J. 1993;12:1311–1319. doi: 10.1002/j.1460-2075.1993.tb05776.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Landsman D, Bustin M. A signature for the HMG-1 box DNA-binding proteins. Bioessays. 1993;15:539–546. doi: 10.1002/bies.950150807. [DOI] [PubMed] [Google Scholar]
  • 37.Ueda T, Chou H, Kawase T, Shirakawa H, Yoshida M. Acidic C-tail of HMGB1 is required for its target binding to nucleosome linker DNA and transcription stimulation. Biochemistry. 2004;43:9901–9908. doi: 10.1021/bi035975l. [DOI] [PubMed] [Google Scholar]
  • 38.Wang H, Yang H, Czura CJ, Sama AE, Tracey KJ. HMGB1 as a late mediator of lethal systemic inflammation. Am J Respir Crit Care Med. 2001;164:1768–1773. doi: 10.1164/ajrccm.164.10.2106117. [DOI] [PubMed] [Google Scholar]
  • 39.Onate SA, Prendergast P, Wagner JP, Nissen M, Reeves R, Pettijohn DE, Edwards DP. The DNA-bendingprotein HMG-1 enhances progesterone receptor binding to its target DNA sequences. Mol Cell Biol. 1994;14:3376–3391. doi: 10.1128/mcb.14.5.3376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Verrier CS, Roodi N, Yee CJ, Bailey LR, Jensen RA, Bustin M, Parl FF. High-mobility group (HMG) protein HMG-1 and TATA-binding protein-associated factor TAF(II)30 affect estrogen receptor-mediated transcriptional activation. Mol Endocrinol. 1997;11:1009–1019. doi: 10.1210/mend.11.8.9962. [DOI] [PubMed] [Google Scholar]
  • 41.Zhang CC, Krieg S, Shapiro DJ. HMG-1 stimulates estrogen response element binding by estrogen receptorfrom stablytransfected HeLa cells. Mol Endocrinol. 1999;13:632–643. doi: 10.1210/mend.13.4.0264. [DOI] [PubMed] [Google Scholar]
  • 42.Zappavigna V, Falciola L, Helmer-Citterich M, Mavilio F, Bianchi ME. HMG1 interacts with HOX proteins and enhances their DNA binding and transcriptional activation. EMBO J. 1996;15:4981–4991. [PMC free article] [PubMed] [Google Scholar]
  • 43.Sutrias-Grau M, Bianchi ME, Bernues J. High mobility group protein 1 interacts specifically with the core domain of human TATA box-binding protein and interferes with transcription factor IIB within the pre-initiation complex. J Biol Chem. 1999;274:1628–1634. doi: 10.1074/jbc.274.3.1628. [DOI] [PubMed] [Google Scholar]
  • 44.Calogero S, Grassi F, Aguzzi A, Voigtlander T, Ferrier P, Ferrari S, Bianchi ME. The lack of chromosomal protein Hmg1 does not disrupt cell growth but causes lethal hypoglycaemia in newborn mice. Nat Genet. 1999;22:276–280. doi: 10.1038/10338. [DOI] [PubMed] [Google Scholar]
  • 45.Wang H, Bloom O, Zhang M, Vishnubhakat JM, Ombrellino M, Che J, Frazier A, Yang H, Ivanova S, Borovikova L, Manogue KR, Faist E, Abraham E, Andersson J, Andersson U, Molina PE, Abumrad NN, Sama A, Tracey KJ. HMG-1 as a late mediator of endotoxin lethality in mice. Science. 1999;285:248–251. doi: 10.1126/science.285.5425.248. [DOI] [PubMed] [Google Scholar]
  • 46.Li J, Kokkola R, Tabibzadeh S, Yang R, Ochani M, Qiang X, Harris HE, Czura CJ, Wang H, Ulloa L, Wang H, Warren HS, Moldawer LL, Fink MP, Andersson U, Tracey KJ, Yang H. Structural basis for the pro inflammatory cytokine activity of high mobility group box 1. Mol Med. 2003;9:37–45. [PMC free article] [PubMed] [Google Scholar]
  • 47.Messmer D, Yang H, Telusma G, Knoll F, Li J, Messmer B, Tracey KJ, Chiorazzi N. High mobility group box protein 1: an endogenous signal for dendritic cell maturation and Th1 polarization. J Immunol. 2004;173:307–313. doi: 10.4049/jimmunol.173.1.307. [DOI] [PubMed] [Google Scholar]
  • 48.Yang H, Ochani M, Li J, Qiang X, Tanovic M, Harris HE, Susarla SM, Ulloa L, Wang H, Di Raimo R, Czura CJ, Wang H, Roth J, Warren HS, Fink MP, Fenton MJ, Andersson U, Tracey KJ. Reversing established sepsis with antagonists of endogenous high-mobility group box 1. Proc Natl Acad Sci USA. 2004;101:296–301. doi: 10.1073/pnas.2434651100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Dumitriu IE, Baruah P, Manfredi AA, Bianchi ME, Rovere-Querini P. HMGB1: guiding immunity from within. Trends Immunol. 2005;26:381–387. doi: 10.1016/j.it.2005.04.009. [DOI] [PubMed] [Google Scholar]
  • 50.Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 2002;418:191–195. doi: 10.1038/nature00858. [DOI] [PubMed] [Google Scholar]
  • 51.Kazama H, Ricci JE, Herndon JM, Hoppe G, Green DR, Ferguson TA. Induction of immu-nological tolerance by apoptotic cells requires caspase-dependent oxidation of high-mobility group box-1 protein. Immunity. 2008;29:21–32. doi: 10.1016/j.immuni.2008.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bonaldi T, Talamo F, Scaffidi P, Ferrera D, Porto A, Bachi A, Rubartelli A, Agresti A, Bianchi ME. Monocytic cells hyperacetylate chromatin protein HMGB1to redirect it towards secretion. EMBO J. 2003;22:5551–5560. doi: 10.1093/emboj/cdg516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Ulloa L, Messmer D. High-mobility group box 1 (HMGB1) protefriend and foe. Cytokine Growth Factor Rev. 2006;17:189–201. doi: 10.1016/j.cytogfr.2006.01.003. [DOI] [PubMed] [Google Scholar]
  • 54.Hori O, Brett J, Slattery T, Cao R, Zhang J, Chen JX, Nagashima M, Lundh ER, Vijay S, Nitecki D. The receptor for advanced glycation end products (RAGE) is a cellular binding site for amphoterin. Mediation of neurite outgrowth and co-expression of rage and amphoterin in the developing nervous system. J Biol Chem. 1995;270:25752–25761. doi: 10.1074/jbc.270.43.25752. [DOI] [PubMed] [Google Scholar]
  • 55.Chavakis T, Bierhaus A, Nawroth PP. RAGE (receptorfor advanced glycation end products): a central player in the inflammatory response. Microbes Infect. 2004;6:1219–1225. doi: 10.1016/j.micinf.2004.08.004. [DOI] [PubMed] [Google Scholar]
  • 56.Taguchi A, Blood DC, del TG, Canet A, Lee DC, Qu W, Tanji N, Lu Y, Lalla E, Fu C, Hofmann MA, Kislinger T, Ingram M, Lu A, Tanaka H, Hori O, Ogawa S, Stern DM, Schmidt AM. Blockade of RAGE-amphoterin signalling suppresses tumour growth and metastases. Nature. 2000;405:354–360. doi: 10.1038/35012626. [DOI] [PubMed] [Google Scholar]
  • 57.Yang H, Wang H, Czura CJ, Tracey KJ. The cytokine activity of HMGB1. J Leukoc Biol. 2005;78:1–8. doi: 10.1189/jlb.1104648. [DOI] [PubMed] [Google Scholar]
  • 58.Huttunen HJ, Rauvala H. Amphoterin as an extracellular regulator of cell motility: from discovery to disease. J Intern Med. 2004;255:351–366. doi: 10.1111/j.1365-2796.2003.01301.x. [DOI] [PubMed] [Google Scholar]
  • 59.Huttunen HJ, Fages C, Rauvala H. Receptor for advanced glycation end products (RAGE)-mediated neurite outgrowth and activation of NF-kappaB require the cytoplasmic domain of the receptor but different downstream signaling pathways. J Biol Chem. 1999;274:19919–19924. doi: 10.1074/jbc.274.28.19919. [DOI] [PubMed] [Google Scholar]
  • 60.Palumbo R, Galvez BG, Pusterla T, De MF, Cossu G, Marcu KB, Bianchi ME. Cells migrating to sites of tissue damage in response to the danger signal HMGB1 require NF-kappaB activation. J Cell Biol. 2007;179:33–40. doi: 10.1083/jcb.200704015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Park JS, Arcaroli J, Yum HK, Yang H, Wang H, Yang KY, Choe KH, Strassheim D, Pitts TM, Tracey KJ, Abraham E. Activation of gene expression in human neutrophils by high mobility group box 1 protein. Am J Physiol Cell Physiol. 2003;284:C870–C879. doi: 10.1152/ajpcell.00322.2002. [DOI] [PubMed] [Google Scholar]
  • 62.Fiuza C, Bustin M, Talwar S, Tropea M, Gerstenberger E, Shelhamer JH, Suffredini AF. Inflammation-promoting activity of HMGB1 on human microvascular endothelial cells. Blood. 2003;101:2652–2660. doi: 10.1182/blood-2002-05-1300. [DOI] [PubMed] [Google Scholar]
  • 63.Treutiger CJ, Mullins GE, Johansson AS, Rouhiainen A, Rauvala HM, Erlandsson-Harris H, Andersson U, Yang H, Tracey KJ, Andersson J, Palmblad JE. High mobility group 1 B-box mediates activation of human endothelium. J Intern Med. 2003;254:375–385. doi: 10.1046/j.1365-2796.2003.01204.x. [DOI] [PubMed] [Google Scholar]
  • 64.Dumitriu IE, Baruah P, Valentinis B, Voll RE, Herrmann M, Nawroth PP, Arnold B, Bianchi ME, Manfredi AA, Rovere-Querini P. Release of high mobility group box1 by dendritic cells controls T cell activation via the receptor for advanced glycation end products. J Immunol. 2005;174:7506–7515. doi: 10.4049/jimmunol.174.12.7506. [DOI] [PubMed] [Google Scholar]
  • 65.Dumitriu IE, Baruah P, Bianchi ME, Manfredi AA, Rovere-Querini P. Requirement of HMGB1 and RAGE for the maturation of human plas-macytoid dendritic cells. Eur J Immunol. 2005;35:2184–2190. doi: 10.1002/eji.200526066. [DOI] [PubMed] [Google Scholar]
  • 66.Dumitriu IE, Bianchi ME, Bacci M, Manfredi AA, Rovere-Querini P. The secretion of HMGB1 is required for the migration of maturing dendritic cells. J Leukoc Biol. 2007;81:84–91. doi: 10.1189/jlb.0306171. [DOI] [PubMed] [Google Scholar]
  • 67.Telusma G, Datta S, Mihajlov I, Ma W, Li J, Yang H, Newman W, Messmer BT, Minev B, Schmidt-Wolf IG, Tracey KJ, Chiorazzi N, Messmer D. Dendritic cell activating peptides induce distinct cytokine profiles. Int Immunol. 2006;18:1563–1573. doi: 10.1093/intimm/dxl089. [DOI] [PubMed] [Google Scholar]
  • 68.Yang D, Chen Q, Yang H, Tracey KJ, Bustin M, Oppenheim JJ. High mobility group box-1 protein induces the migration and activation of human dendritic cells and acts as an alarm-in. J Leukoc Biol. 2007;81:59–66. doi: 10.1189/jlb.0306180. [DOI] [PubMed] [Google Scholar]
  • 69.Orlova VV, Choi EY, Xie C, Chavakis E, Bierhaus A, Ihanus E, Ballantyne CM, Gahmberg CG, Bianchi ME, Nawroth PP, Chavakis T. A novel pathway of HMGB1-mediated inflammatory cell recruitment that requires Mac-1-integrin. EMBO J. 2007;26:1129–1139. doi: 10.1038/sj.emboj.7601552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Rouhiainen A, Kuja-Panula J, Wilkman E, Pakkanen J, Stenfors J, Tuominen RK, Lepantalo M, Carpen O, Parkkinen J, Rauvala H. Regulation of monocyte migration by ampho-terin (HMGB1) Blood. 2004;104:1174–1182. doi: 10.1182/blood-2003-10-3536. [DOI] [PubMed] [Google Scholar]
  • 71.Yu M, Wang H, Ding A, Golenbock DT, Latz E, Czura CJ, Fenton MJ, Tracey KJ, Yang H. HMGB1 signals through toll-like receptor (TLR) 4 and TLR2. Shock. 2006;26:174–179. doi: 10.1097/01.shk.0000225404.51320.82. [DOI] [PubMed] [Google Scholar]
  • 72.Park JS, Gamboni-Robertson F, He Q, Svetkauskaite D, Kim JY, Strassheim D, Sohn JW, Yamada S, Maruyama I, Banerjee A, Ishizaka A, Abraham E. High mobility group box 1 protein interacts with multiple Toll-like receptors. Am J Physiol Cell Physiol. 2006;290:C917–C924. doi: 10.1152/ajpcell.00401.2005. [DOI] [PubMed] [Google Scholar]
  • 73.Park JS, Svetkauskaite D, He Q, Kim JY, Strassheim D, Ishizaka A, Abraham E. Involvement of toll-like receptors 2 and 4 in cellular activation by high mobility group box 1 protein. J Biol Chem. 2004;279:7370–7377. doi: 10.1074/jbc.M306793200. [DOI] [PubMed] [Google Scholar]
  • 74.Faure E, Thomas L, Xu H, Medvedev A, Equils O, Arditi M. Bacterial lipopolysaccharide and IFN-gamma induce Toll-like receptor 2 and Toll-like receptor 4 expression in human endothelial cells: role of NF-kappa B activation. J Immunol. 2001;166:2018–2024. doi: 10.4049/jimmunol.166.3.2018. [DOI] [PubMed] [Google Scholar]
  • 75.Underhill DM, Ozinsky A, Smith KD, Aderem A. Toll-like receptor-2 mediates mycobacteria-induced proinflammatory signaling in macrophages. Proc Natl Acad Sci USA. 1999;96:14459–14463. doi: 10.1073/pnas.96.25.14459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Kawai T, Akira S. Signaling to NF-kappaB by Toll-like receptors. Trends Mol Med. 2007;13:460–469. doi: 10.1016/j.molmed.2007.09.002. [DOI] [PubMed] [Google Scholar]
  • 77.Ivanov S, Dragoi AM, Wang X, Dallacosta C, Louten J, Musco G, Sitia G, Yap GS, Wan Y, Biron CA, Bianchi ME, Wang H, Chu WM. A novel role for HMGB1 in TLR9-mediated inflammatory responses to CpG-DNA. Blood. 2007;110:1970–1981. doi: 10.1182/blood-2006-09-044776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Brilot F, Geenen V. [Role of viral infections in the pathogenesis of type 1 diabetes] Rev Med Liege. 2005;60:297–302. [PubMed] [Google Scholar]
  • 79.Gillespie KM. Type 1 diabetes: pathogenesis and prevention. CMAJ. 2006;175:165–170. doi: 10.1503/cmaj.060244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Von Herrath MG. Pathogenesis of type 1 diabetes: a viewpoint. Adv Exp Med Biol. 2004;552:317–321. [PubMed] [Google Scholar]
  • 81.Knip M, Veijola R, Virtanen SM, Hyoty H, Vaarala O, Akerblom HK. Environmental triggers and determinants of type 1 diabetes. Diabetes. 2005;54(Suppl 2):S125–S136. doi: 10.2337/diabetes.54.suppl_2.s125. [DOI] [PubMed] [Google Scholar]
  • 82.Ziegler AG, Standl E. [Type-1 diabetes: the pathogenesis of an immune disease] Dtsch Med Wochenschr. 1994;119:705–706. doi: 10.1055/s-0029-1235055. [DOI] [PubMed] [Google Scholar]
  • 83.Scherbaum WA. Etiology and pathogenesis of type 1 diabetes. Horm Metab Res Suppl. 1992;26:111–116. [PubMed] [Google Scholar]
  • 84.Jansen A, Homo-Delarche F, Hooijkaas H, Leenen PJ, Dardenne M, Drexhage HA. Immunohistochemical characterization of monocytes-macrophages and dendritic cells involved in the initiation of the insulitis and beta-cell destruction in NOD mice. Diabetes. 1994;43:667–675. doi: 10.2337/diab.43.5.667. [DOI] [PubMed] [Google Scholar]
  • 85.Charre S, Rosmalen JG, Pelegri C, Alves V, Leenen PJ, Drexhage HA, Homo-Delarche F. Abnormalities in dendritic cell and macrophage accumulation in the pancreas of nonobese diabetic (NOD) mice during the early neonatal period. Histol Histopathol. 2002;17:393–401. doi: 10.14670/HH-17.393. [DOI] [PubMed] [Google Scholar]
  • 86.Grodzicky T, Elkon KB. Apoptosis: a case where too much or too little can lead to autoimmunity. MtSinai J Med. 2002;69:208–219. [PubMed] [Google Scholar]
  • 87.Kalden J. Apoptosis in systemic autoimmunity. Autoimmun Rev. 2004;3(Suppl 1):S9–10. [PubMed] [Google Scholar]
  • 88.Manfredi AA, Sabbadini MG, Rovere-Querini P. Dendritic cells and the shadow line between autoimmunity and disease. Arthritis Rheum. 2005;52:11–15. doi: 10.1002/art.20758. [DOI] [PubMed] [Google Scholar]
  • 89.Pisetsky DS. The immune response to cell death in SLE. Autoimmun Rev. 2004;3:500–504. doi: 10.1016/j.autrev.2004.07.010. [DOI] [PubMed] [Google Scholar]
  • 90.Scaglia L, Cahill CJ, Finegood DT, Bonner-Weir S. Apoptosis participates in the remodeling of the endocrine pancreas in the neonatal rat. Endocrinology. 1997;138:1736–1741. doi: 10.1210/endo.138.4.5069. [DOI] [PubMed] [Google Scholar]
  • 91.Sheriff A, Gaipl US, Voll RE, Kaden JR, Herrmann M. Apoptosis and systemic lupus erythematosus. Rheum Dis Clin North Am. 2004;30:505–5ix. doi: 10.1016/j.rdc.2004.04.006. [DOI] [PubMed] [Google Scholar]
  • 92.Trudeau JD, Dutz JP, Arany E, Hill DJ, Fieldus WE, Finegood DT. Neonatal beta-cell apoptosis: a trigger for autoimmune diabetes? Diabetes. 2000;49:1–7. doi: 10.2337/diabetes.49.1.1. [DOI] [PubMed] [Google Scholar]
  • 93.Turley S, Poirot L, Hattori M, Benoist C, Mathis D. Physiological beta cell death triggers priming of self-reactive T cells by dendritic cells in a type-1 diabetes model. J Exp Med. 2003;198:1527–1537. doi: 10.1084/jem.20030966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Gaipl US, Voll RE, Sheriff A, Franz S, Kalden JR, Herrmann M. Impaired clearance of dying cells in systemic lupus erythematosus. Autoimmun Rev. 2005;4:189–194. doi: 10.1016/j.autrev.2004.10.007. [DOI] [PubMed] [Google Scholar]
  • 95.Shimaoka Y, Attrep JF, Hirano T, Ishihara K, Suzuki R, Toyosaki T, Ochi T, Lipsky PE. Nurse-like cells from bone marrow and synovium of patients with rheumatoid arthritis promote survival and enhance function of human B cells. J Clin Invest. 1998;102:606–618. doi: 10.1172/JCI3162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Fujisawa K, Asahara H, Okamoto K, Aono H, Hasunuma T, Kobata T, Iwakura Y, Yonehara S, Sumida T, Nishioka K. Therapeutic effect of the anti-Fas antibody on arthritis in HTLV-1 tax transgenic mice. J Clin Invest. 1996;98:271–278. doi: 10.1172/JCI118789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Sabelko KA, Kelly KA, Nahm MH, Cross AH, Russell JH. Fas and Fas ligand enhance the pathogenesis of experimental allergic encephalomyelitis, but are not essential for immune privilege in the central nervous system. J Immunol. 1997;159:3096–3099. [PubMed] [Google Scholar]
  • 98.Waldner H, Sobel RA, Howard E, Kuchroo VK. Fas- and FasL-deficient mice are resistant to induction of autoimmune encephalomyelitis. J Immunol. 1997;159:3100–3103. [PubMed] [Google Scholar]
  • 99.Bell CW, Jiang W, Reich CF, III, Pisetsky DS. The extracellular release of HMGB1 during apop-totic cell death. Am J Physiol Cell Physiol. 2006;291:C1318–C1325. doi: 10.1152/ajpcell.00616.2005. [DOI] [PubMed] [Google Scholar]
  • 100.Qin S, Wang H, Yuan R, Li H, Ochani M, Ochani K, Rosas-Ballina M, Czura CJ, Huston JM, Miller E, Lin X, Sherry B, Kumar A, Larosa G, Newman W, Tracey KJ, Yang H. Role of HMGB1 in apoptosis-mediated sepsis lethality. J Exp Med. 2006;203:1637–1642. doi: 10.1084/jem.20052203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Choi JJ, Reich CF, III, Pisetsky DS. The role of macrophages in the in vitro generation of extracellular DNA from apoptotic and necrotic cells. Immunology. 2005;115:55–62. doi: 10.1111/j.1365-2567.2005.02130.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Choi JJ, Reich CF, III, Pisetsky DS. Release of DNA from dead and dying lymphocyte and monocyte cell lines in vitro. Scand J Immunol. 2004;60:159–166. doi: 10.1111/j.0300-9475.2004.01470.x. [DOI] [PubMed] [Google Scholar]
  • 103.Han J, Zhong J, Wei W, Wang Y, Huang Y, Yang P, Purohit S, Dong Z, Wang MH, She JX, Gong F, Stern DM, Wang CY. Extracellular high-mobility group box 1 acts as an innate immune mediator to enhance autoimmune progression and diabetes onset in NOD mice. Diabetes. 2008;57:2118–2127. doi: 10.2337/db07-1499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.O'Brien BA, Huang Y, Geng X, Dutz JP, Finegood DT. Phagocytosis of apoptotic cells by macrophages from NOD mice is reduced. Diabetes. 2002;51:2481–2488. doi: 10.2337/diabetes.51.8.2481. [DOI] [PubMed] [Google Scholar]
  • 105.O'Brien BA, Fieldus WE, Field CJ, Finegood DT. Clearance of apoptotic beta-cells is reduced in neonatal autoimmune diabetes-prone rats. Cell Death Differ. 2002;9:457–464. doi: 10.1038/sj.cdd.4400973. [DOI] [PubMed] [Google Scholar]
  • 106.O'Brien BA, Geng X, Orteu CH, Huang Y, Ghoreishi M, Zhang Y, Bush JA, Li G, Finegood DT, Dutz JP. A deficiency in the in vivo clearance of apoptotic cells is a feature of the NOD mouse. J Autoimmun. 2006;26:104–115. doi: 10.1016/j.jaut.2005.11.006. [DOI] [PubMed] [Google Scholar]
  • 107.Mathis D, Vence L, Benoist C. beta-Cell death during progression to diabetes. Nature. 2001;414:792–798. doi: 10.1038/414792a. [DOI] [PubMed] [Google Scholar]
  • 108.Jaeschke H, Lemasters JJ. Apoptosis versus on-cotic necrosis in hepatic ischemia/reperfusion injury. Gastroenterology. 2003;125:1246–1257. doi: 10.1016/s0016-5085(03)01209-5. [DOI] [PubMed] [Google Scholar]
  • 109.Atkinson MA, Eisenbarth GS. Type 1 diabetes: new perspectives on disease pathogenesis and treatment. Lancet. 2001;358:221–229. doi: 10.1016/S0140-6736(01)05415-0. [DOI] [PubMed] [Google Scholar]
  • 110.Andre I, Gonzalez A, Wang B, Katz J, Benoist C, Mathis D. Checkpoints in the progression of autoimmune disease: lessons from diabetes models. Proc Natl Acad Sci USA. 1996;93:2260–2263. doi: 10.1073/pnas.93.6.2260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Vasquez AC, Feili-Hariri M, Tan RJ, Morel PA. Qualitative and quantitative abnormalities in splenic dendritic cell populations in NOD mice. Clin Exp Immunol. 2004;135:209–218. doi: 10.1111/j.1365-2249.2003.02359.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Gruessner AC, Sutherland DE. Pancreas transplant outcomes for United States (US) and non-US cases as reported to the United Network for Organ Sharing (UNOS) and the International Pancreas Transplant Registry (IPTR) as of June 2004. Clin Transplant. 2005;19:433–455. doi: 10.1111/j.1399-0012.2005.00378.x. [DOI] [PubMed] [Google Scholar]
  • 113.Burke GW, Ciancio G, Sollinger HW. Advances in pancreas transplantation. Transplantation. 2004;77:S62–S67. doi: 10.1097/01.tp.0000126929.71923.77. [DOI] [PubMed] [Google Scholar]
  • 114.Ryan EA, Paty BW, Senior PA, Bigam D, Alfadhli E, Kneteman NM, Lakey JR, Shapiro AM. Five-year follow-up after clinical islet transplantation. Diabetes. 2005;54:2060–2069. doi: 10.2337/diabetes.54.7.2060. [DOI] [PubMed] [Google Scholar]
  • 115.Shapiro AM, Lakey JR, Ryan EA, Korbutt GS, Toth E, Warnock GL, Kneteman NM, Rajotte RV. Islet transplantation in seven patients with type 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N Engl J Med. 2000;343:230–238. doi: 10.1056/NEJM200007273430401. [DOI] [PubMed] [Google Scholar]
  • 116.Shapiro AM, Ricordi C, Hering BJ, Auchincloss H, Lindblad R, Robertson RP, Secchi A, Brendel MD, Berney T, Brennan DC, Cagliero E, Alejandro R, Ryan EA, Di Mercurio B, Morel P, Polonsky KS, Reems JA, Bretzel RG, Bertuzzi F, Froud T, Kandaswamy R, Sutherland DE, Eisenbarth G, Segal M, Preiksaitis J, Korbutt GS, Barton FB, Viviano L, Seyfert-Margolis V, Bluestone J, Lakey JR. International trial of the Edmonton protocol for islet transplantation. N Engl J Med. 2006;355:1318–1330. doi: 10.1056/NEJMoa061267. [DOI] [PubMed] [Google Scholar]
  • 117.Huang Y, Yin H, Han J, Huang B, Xu J, Zheng F, Tan Z, Fang M, Rui L, Chen D, Wang S, Zheng X, Wang CY, Gong F. Extracellular hmgb1 functions as an innate immune-mediator implicated in murine cardiac allograft acute rejection. Am J Transplant. 2007;7:799–808. doi: 10.1111/j.1600-6143.2007.01734.x. [DOI] [PubMed] [Google Scholar]
  • 118.Lai Y, Chen C, Linn T. Innate immunity and heat shock response in islet transplantation. Clin Exp Immunol. 2009 doi: 10.1111/j.1365-2249.2009.03899.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Tsung A, Sahai R, Tanaka H, Nakao A, Fink MP, Lotze MT, Yang H, Li J, Tracey KJ, Geller DA, Billiar TR. The nuclearfactor HMGB1 mediates hepatic injury after murine liver ischemia-reperfusion. J Exp Med. 2005;201:1135–1143. doi: 10.1084/jem.20042614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Moser B, Szabolcs MJ, Ankersmit HJ, Lu Y, Qu W, Weinberg A, Herold KC, Schmidt AM. Blockade of RAGE suppresses alloimmune reactions in vitro and delays allograft rejection in murine heart transplantation. Am J Transplant. 2007;7:293–302. doi: 10.1111/j.1600-6143.2006.01617.x. [DOI] [PubMed] [Google Scholar]
  • 121.Miller DM, Thornley T, Pearson T, Yamazaki M, Brehm MA, Rossini AA, Greiner DL. TLR agonists abrogate co-stimulation blockade-induced mixed chimerism and transplantation tolerance. Ann N Y Acad Sci. 2008;1150:149–151. doi: 10.1196/annals.1447.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Chen L, Wang T, Zhou P, Ma L, Yin D, Shen J, Molinero L, Nozaki T, Phillips T, Uematsu S, Akira S, Wang CR, Fairchild RL, Alegre ML, Chong A. TLR engagement prevents transplantation tolerance. Am J Transplant. 2006;6:2282–2291. doi: 10.1111/j.1600-6143.2006.01489.x. [DOI] [PubMed] [Google Scholar]
  • 123.Yi S, Wang Y, Chandra AP, O'Hara JM, Wu J, Ouyang L, Burgess JS, Hawthorne W, Wu H, Chadban SJ, O'Connell PJ. Requirement of MyD88 for macrophage-mediated islet xeno-graft rejection after adoptive transfer. Transplantation. 2007;83:615–623. doi: 10.1097/01.tp.0000253759.87886.39. [DOI] [PubMed] [Google Scholar]
  • 124.Goldberg A, Parolini M, Chin BY, Czismadia E, Otterbein LE, Bach FH, Wang H. Toll-like receptor 4 suppression leads to isletallograft survival. FASEBJ. 2007;21:2840–2848. doi: 10.1096/fj.06-7910com. [DOI] [PubMed] [Google Scholar]
  • 125.Walker WE, Nasr IW, Camirand G, Tesar BM, Booth CJ, Goldstein DR. Absence of innate MyD88 signaling promotes inducible allograft acceptance. J Immunol. 2006;177:5307–5316. doi: 10.4049/jimmunol.177.8.5307. [DOI] [PubMed] [Google Scholar]
  • 126.Chandra AP, Ouyang L, Yi S, Wong JK, Ha H, Walters SN, Patel AT, Stokes R, Jardine M, Hawthorne WJ, O'Connell PJ. Chemokine and toll-like receptor signaling in macrophage mediated islet xenograft rejection. Xenotransplantation. 2007;14:48–59. doi: 10.1111/j.1399-3089.2006.00363.x. [DOI] [PubMed] [Google Scholar]
  • 127.Chen Y, Akirav EM, Chen W, Henegariu O, Moser B, Desai D, Shen JM, Webster JC, Andrews RC, Mjalli AM, Rothlein R, Schmidt AM, Clynes R, Herold KC. RAGE ligation affects T cell activation and controls T cell differentiation. J Immunol. 2008;181:4272–4278. doi: 10.4049/jimmunol.181.6.4272. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from International Journal of Clinical and Experimental Pathology are provided here courtesy of e-Century Publishing Corporation

RESOURCES