Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of exocrine pancreatic cancer with a 9% five-year survival rate. High mobility group box 1 (HMGB1) is a nuclear protein that can act as a DNA chaperone in the sustainment of chromosome structure and function. When released into the extracellular space, HMGB1 becomes the most well-characterized damage-associated molecular pattern (DAMP) to trigger immune responses. Recent evidence indicates that intracellular HMGB1 is a novel tumor suppressor in PDAC, which is connected to its role in the prevention of oxidative stress, genomic instability, and histone release. However, since extracellular HMGB1 is a DAMP and pro-inflammatory cytokine, cancer cells can also exploit it to survive through the receptor for advanced glycation endproducts (RAGE) in the pancreatic tumor microenvironment. Interestingly, targeting the HMGB1-RAGE pathway has become a new anticancer therapy strategy for PDAC.
Introduction
Pancreatic ductal adenocarcinoma (PDAC) is the most common histological type of pancreatic cancer, accounting for more than 90% of cases. Pancreatic cancer is the third leading cause of cancer-related death, with a five-year survival rate of just 9% in the United States according to the American Cancer Society (1). In 2018, about 55,440 and 44,330 people will be diagnosed or die with pancreatic cancer in the United States, respectively (1). Indeed, rates are rising faster than any other cancer and the disease is anticipated to become the second leading cause of cancer-related death around 2020 in the United States. Age, smoking, being overweight, pancreatitis, and diabetes are the major known risk factors for pancreatic cancer. Compared to other tumor types, PDAC has different biological features that make it so deadly. The location of the pancreas is deep in the abdominal cavity and pancreatic cancer is seldom detected early or show early noticeable symptoms. In turn, 80% of PDAC patients are diagnosed with advanced metastatic disease, and surgical removal of the tumor is possible in less than 20% of patients with PDAC. In addition, chemotherapy or radiation is commonly used before or after surgery in the treatment of metastatic PDAC (2). Improving knowledge of histology and the genetics of pancreatic tumorigeneses is essential for designing new anticancer approaches to treat this deadly disease (3, 4).
Chromatin, a mass of genetic material composed of proteins, DNA, and RNA, is found inside the nucleus of eukaryotic cells. In 1973, “high mobility group (HMG)” proteins, which can recognize and bind different DNA structures, were first isolated from the chromatin of calf thymus (5). Currently, the HMG family is divided into three superfamilies, namely HMGB (e.g., HMGB1, HMGB2, HMGB3, and HMGB4), HMGA (e.g., HMGA1 and HMGA2), and HMGN (e.g., HMGN1 and HMGN2) according to the characteristic sequence motif (6). Among them, high mobility group box 1 (HMGB1, also termed amphoterin) is a well-characterized non-histone nuclear protein that can be released by various cells under stresses. Dysfunction of HMGB1 signaling contributes to each hallmark of cancer (7, 8). In this review, we outline the basic function of HMGB1 inside and outside of cells and highlight the multifaceted role of HMGB1 in pancreatic tumorigenesis and therapy.
Nuclear HMGB1
HMGB1 was extremely conserved during evolution and contains two HMG boxes that bind and bend DNA. Nuclear HMGB1 is an essential chromosomal architectural factor and implicated in a number of nuclear events such as nucleosome stability and sliding (9). Nucleosome is a basic unit of DNA packaging in eukaryotes, consisting of a segment of DNA wound in sequence around a histone octamer core (histones H2A, H2B, H3, and H4). HMGB1 is required for the assembly of chromatin in vitro (10). HMGB1 can bind to linker histones (H1 and H5) to relax nucleosome, which leads to more accessibility of chromatin at the distorted site. Moreover, loss of HMGB1 decreases the number of nucleosomes by 20–30%, increases histone release, and causes telomere shortening, which in turn affects genome chromatinization (10). HMGB1 has powerful functions in DNA binding, with structure specificity, but not sequence specificity (11). HMGB1 not only binds to normal DNA, but also to distorted and damaged DNA such as H-DNA (12), four-way DNA junctions (13–15), and oxidative-damaged DNA (16). As a DNA chaperone, HMGB1 can bend and change DNA structure and morphology by unwinding (17), looping (18), or compacting DNA (17). This DNA chaperone activity of HMGB1 contributes to V(D)J recombination, gene transcription, DNA replication, DNA repair, and gene transfer. Consequently, loss of HMGB1 increases DNA damage and decreases DNA repair efficiency in response to oxidative stress and injury (19).
Cytosolic HMGB1
It is well known that most of HMGB1 is found in the nucleus, whereas little HMGB1 has been observed in the cytoplasm under normal conditions. In contrast, HMGB1 translocates from the nucleus to the cytosol and then releases into the extracellular space under various stresses, especially inflammatory stimuli and oxidative stress (20). Although the function of cytosolic HMGB1 remains largely unknown, it plays a potential role in promoting autophagy (21). Autophagy is a lysosomal-dependent degradation pathway that removes unused proteins or damaged organelles. Disruption of autophagic pathways is implicated in multiple human diseases including neurodegenerative diseases, cancer, and inflammatory diseases. Cytosolic HMGB1 promotes autophagy through directly binding to BECN1 (also termed ATG6), a core component of the class III phosphatidylinositol 3-kinase complexes (21). HMGB1 is a redox protein and contains three conserved redox-sensitive cysteine residues: C23 and C45 can form an intramolecular disulfide bond, whereas C106 affects HMGB1 localization. Mutation of cysteine 106 (C106) increases cytosolic HMGB1 and subsequent interaction between HMGB1 and BECN1 in the induction of autophagy (21). Nuclear and extracellular HMGB1 also promotes autophagy, which will be discussed later. In addition, cytosolic HMGB1 is involved in the regulation of an unconventional secretory pathway in the lysosome through binding to annexin A2, myosin-9, and Rab10 (22). It remains unclear whether HMGB1-dependent autophagy is required for HMGB1-mediated protein secretion.
Extracellular HMGB1
In 1999, Dr. Haichao Wang’s lab reported that HMGB1 is secreted into the extracellular space by activated macrophages and functions as a late lethal mediator in pathogen infection (23). In addition to its active secretion by immune cells, HMGB1 can be passively released by dead, dying, or injured cells as a DAMP in sterile inflammation (24). Indeed, HMGB1 release is observed in various types of cell death including necrosis, necroptosis, apoptosis, ferroptosis, pyroptosis, and autophagic cell death (25). Although the release mechanism may change depending on the cell death type, oxidative stress plays a central role in the coordination of cell death and HMGB1 release (26). Once released, extracellular HMGB1 can trigger an inflammatory and immune response through both receptor-dependent and -independent manners. A number of receptors have been identified to mediate extracellular HMGB1 activity in vitro or in vivo. In particular, Toll like receptor (TLR) and the receptor for advanced glycation endproducts (RAGE) play a major role in the control of HMGB1 activity in immune cells such as macrophages (27), monocytes (28), neutrophils (29, 30), eosinophils, astrocytes (31), fibroblasts (32–34), keratinocytes (35), dendritic cells (36), natural killer cells, and T cells. Exogenous HMGB1 promotes cell migration, invasion, and proliferation through activation of several signaling pathways such as the mitogen-activated protein kinase pathway, the NF-κB pathway, and the PI3K/AKT/mTOR pathway (25). Apart from direct receptor interaction, HMGB1 may form heterocomplexes with other immune co-activators such as IL-1, CXCL12, DNA, nucleosome, or lipopolysaccharide that generate synergistic responses in inflammation and immunity (37).
HMGB1 and acute pancreatitis
As a basic pathological process of various diseases, inflammation contributes to progressive pancreatic disease ranging from pancreatitis to pancreatic cancer. Acute pancreatitis (AP) is an inflammatory process of the pancreatic gland that exhibits a broad clinical spectrum and its severity may vary from mild and edematous to a serious, necrotizing disease with high morbidity and mortality (38). In its most severe forms, AP involves remote organ systems. In fact, systemic inflammatory response syndrome (SIRS) is one of the major pathobiological processes underlying severe AP. It is widely accepted that the premature activation of digestive enzymes (trypsin, elastase, and lipase) within pancreatic acinar cells is a critical initiating event that leads to organelle injury and autodigestion of the pancreas (39). However, AP is also an inflammatory disorder involving a complex cascade of immunological events, including inflammatory mediator production, which affects not only the pathogenesis but also the course of the disease. Some of these inflammatory mediators are initially released by pancreatic acinar cells and result in the recruitment and activation of neutrophils, monocytes, and macrophages (40–43), which lead to further acinar cell injury. When released, these mediators gain access to the systemic circulation and play a central role in the progression of SIRS and multisystem organ failure (44). However, the exact molecular mechanisms linking the progression of local pancreatic damage to systemic inflammation are still poorly understood.
Serum levels of HMGB1 were significantly elevated in patients with AP and were correlated with the severity of the disease (45). Early blockade or delayed therapeutic delivery (e.g., ethyl pyruvate (46), A box (47) and anti-HMGB1 neutralizing antibody (48)) targeting HMGB1 significantly attenuates the development and associated organ dysfunction in experimental AP. In addition, antioxidant (e.g., pyrrolidine dithiocarbamate (49)) and anticoagulant (e.g., antithrombin III (50) and danaparoid sodium (51)) inhibits HMGB1 release and reduce pancreatic injury in severe AP. These findings indicate that extracellular HMGB1 mediates the inflammatory response and may be an effective therapeutic target of AP. In contrast, intracellular HMGB1 protects against AP (52). Conditional knockout of HMGB1 in the pancreas fails to affect pancreatic development and function (52). However, HMGB1 depletion in the pancreas increases animal death in mice following administration of L-arginine or cerulean (52). Loss of endogenous HMGB1 in the pancreas increases DNA damage and subsequent cell death, nuclear DAMP release (e.g., histone and DNA), and the inflammatory response (52). Moreover, histone and DNA can activate macrophages to release HMGB1, which amplifies the inflammation response in AP (52). These findings may explain increased DNA damage and HMGB1 release in the loss of pancreatic HMGB1.
HMGB1 and pancreatic tumorigenesis
PDAC is characterized by a high frequency (>95%) of activation of K-Ras mutations (especially G12D mutation) (53, 54) and progresses from non-invasive pancreatic lesions that include pancreatic intraepithelial neoplasias (PanINs), intraductal papillary mucinous neoplasms (IPMNs), and mucinous cystic neoplasms (MCNs) (55). The ability of mutant K-Ras to drive PDAC was not successfully investigated until the generation of mice with a Cre-inducible conditional allele (Pdx1-Cre;K-RasG12D/+, termed KC mice) targeting the endogenous K-Ras locus (56). These KC mice develop lesions that slowly progress further into advanced PDAC and have a median survival of 15 months (56), suggesting that K-Ras activation is a tumor-initiating event that requires other elements that accelerate rigorous PDAC progression. We recently developed a novel mouse model combining original K-RasG12D/+-driven PDAC models with additional loss of HMGB1 in pancreatic tissue (Pdx1-Cre;K-RasG12D/+;HMGB1−/−, termed KCH mice). Our data demonstrated that intracellular HMGB1 is a novel tumor suppressor of PDAC by sustaining chromosome stability and limiting pro-inflammatory nucleosome release and activity in mice (57).
Epidemiological studies have established a significant correlation between pancreatic cancer and diabetes. People with type 2 diabetes are well known to be at increased risk for pancreatic cancer, and now it seems that the risk extends to those with type 1 diabetes (58). The risk of developing pancreatic cancer was twice as high in subjects with type 1 or young-onset diabetes as that in people without diabetes (59). Diabetes is either a risk factor or a symptom of pancreatic cancer (58, 60–62). Much is known about these associations, but why pancreatic cancer causes diabetes and how diabetes affects the outcome of pancreatic cancer have yet to be fully determined. Remarkably, 100% of KCH mice also develop type 1 diabetes at five-eight weeks and have an average survival rate of three months (57). Importantly, PDAC patients had reduced pancreatic HMGB1 expression with poor survival outcomes (57). In addition, diabetic human and mouse tissues contain lower levels of HMGB1 expression than their normoglycemic counterparts (63). These findings suggest that loss of HMGB1 expression plays an important pathogenic role in human pancreatic cancer and diabetes.
The emerging role of autophagy in pancreatic cancer and diabetes
Autophagy is an evolutionarily-conserved degradation pathway by which cytoplasmic components, including damaged organelles (e.g., mitochondria or endoplasmic reticulum [ER]) and effete long-lived proteins are digested within the lysosome (64, 65). This dynamic process is primarily controlled by members of the autophagy-related gene (ATG) family and share regulators derived from other trafficking and cell death pathways. The role of autophagy in cancer is complex and is likely dependent on tumor type, stage, genetic context, and tumor microenvironment (66). On one hand, autophagy acts as a tumor suppressor at an early stage by preventing genome instability, limiting oxidative stress, reducing intratumoral necrosis-dependent inflammation, and inhibiting angiogenesis. On the other hand, autophagy acts as a survival mechanism at a late stage that can promote the growth of established tumors and resistance to anticancer treatment by providing substrates for metabolism, removing damaged organelles and proteins, and diminishing apoptosis. Recent studies demonstrated that mice lacking the essential autophagy genes (e.g., ATG5 or ATG7) or mitophagy genes (e.g., PINK1 or PARK2) have accelerated K-Ras-driven PanIN formation and reduced animal survival during tumorigenesis (67, 68). In contrast, increased autophagy promotes pancreatic growth at the late stage (69, 70). In addition, increasing evidence supports an active role for autophagy in the pathophysiology of type 1 and type 2 diabetes (71). Increased autophagy is necessary to maintain the mass and function of pancreatic β cells and protects β cells against damage by oxidative stress in diabetic mice (72, 73). Thus, β-cell-specific ATG7-null mice show hypoinsulinemia and hyperglycemia (74). These studies support the idea that autophagy plays an important pathogenic role in the regulation of pancreatic tumor and diabetes development.
HMGB1 as a novel regulator of autophagy
Our studies have demonstrated that HMGB1 has transcription-dependent and transcription-independent pro-autophagic functions. Heat shock protein beta-1 (HSPB1), a regulator of actin cytoskeleton dynamics, is a direct transcriptional target of HMGB1 in autophagy (75). Suppression of HSPB1 and HMGB1 expression inhibits the dynamics of autophagy as well as mitophagy, a form of selective autophagy that removes damaged mitochondria (75). During stress, including starvation, oxidative stress, and chemotherapy, HMGB1 translocates from the nucleus to the cytoplasm and binds to Beclin-1, which initiates autophagosome formation (21). p53 and unc-51-like kinase 1 (ULK1, also called ATG1 in yeast) have opposing roles in the regulation of HMGB1-Beclin-1 complex formation in cancer cells (76, 77). The ULK1 kinase is an essential component of the core autophagy machinery that regulates autophagosome formation. Once released, reduced HMGB1 triggers autophagy in a RAGE-dependent manner in cancer cells, which promotes tumor cell proliferation (78). In contrast, HMGB1 may not be required for autophagy in some organs such as the liver and heart (79). Thus, understanding HMGB1-dependent and -independent autophagy in more detail will provide insight into the integrated stress response and guide HMGB1-based therapeutic intervention in cancer and other diseases (80).
RAGE as a critical receptor of nuclear danger signal in pancreatic tumorigenesis
RAGE is a transmembrane receptor of the immunoglobulin gene superfamily and a multifunctional receptor within the tumor microenvironment. RAGE expression is associated with inflammation and is implicated in several chronic diseases, including cancer (81, 82). RAGE and its ligands are linked to the development and progression of several cancers by facilitating the maintenance of a chronic inflammatory state (83) and/or by promotion of metastases (84). RAGE is expressed by cancer cells as well as other cells within the tumor microenvironment, including T cells (85), macrophages (86), endothelial cells (87), and fibroblasts (88). We recently provided the first evidence that RAGE plays a unique role in pancreatic tumorigenesis and drug resistance in vitro and in vivo (89–93). We demonstrated that: 1) RAGE was highly expressed in mouse and human PDAC (90); 2) Targeted genetic ablation of RAGE in mice prevented pancreatic cancer growth in a genetically-modified spontaneous mouse model (Pdx1-Cre;K-Ras G12D/+;RAGE−/−, termed KCR mice; B6 background) (90) and a xenograft mouse model (93); 3) RAGE was essential for oncogenic K-Ras-mediated hypoxic signaling in pancreatic cancer development (94); and 4) The mechanism by which this occurs in part involves inflammatory response-associated metabolic changes (90, 93, 95), cell death-promoting limitations in autophagy (91, 92, 96, 97), and a reduction in the accumulation of MDSCs and Tregs (89). In addition, RAGE (but not TLR9) deletion limits pancreatic cancer development in KCH mice (57). These exciting findings indicate that RAGE plays an important role in the pathogenesis of pancreatic cancer.
Conclusions and Perspectives
PDAC is driven by mutant oncogenic K-Ras and has a lower overall five-year survival rate. Numerous trials have failed to improve outcomes of this deadliest of all major cancers; potential causes of this failure include a still insufficient understanding of PDAC’s key features and imperfect preclinical models for identification of active agents and mechanisms of therapeutic responses and resistance. HMGB1, a highly-conserved chromosomal protein, plays an important role in human diseases, including PDAC. Interestingly, intracellular HMGB1 suppresses whereas extracellular HMGB1 promotes pancreatic tumorigenesis. HMGB1 translocation from the nucleus or deficiency-mediated nucleosome release seem to be key molecular events linking chromosomal instability and the inflammatory response. This process requires activation of RAGE, a DAMP receptor that promotes inflammatory responses to HMGB1 (93), DNA (98), and histones (99). Importantly, pharmacological inhibition of intracellular HMGB1 loss or release by glycyrrhizin limits K-Ras-driven tumorigenesis in mice. Information gleaned from such preclinical studies need to be further studied in the clinic to impact PDAC detection and treatment.
Acknowledgments
We thank Christine Burr (Department of Surgery, University of Pittsburgh) for her critical reading of the manuscript. This work was supported by grants from the US National Institutes of Health (R01GM115366, R01CA160417, and R01CA211070), the Natural Science Foundation of Guangdong Province (2016A030308011), the American Cancer Society (Research Scholar Grant RSG-16-014-01-CDD), the National Natural Science Foundation of China (31671435, 81400132, and 81772508), and Guangdong Province Universities and Colleges Pearl River Scholar Funded Scheme (2017).
References
- 1.Siegel RL, Miller KD, and Jemal A 2018. Cancer statistics, 2018. CA Cancer J Clin 68: 7–30. [DOI] [PubMed] [Google Scholar]
- 2.Garrido-Laguna I, and Hidalgo M 2015. Pancreatic cancer: from state-of-the-art treatments to promising novel therapies. Nature reviews. Clinical oncology 12: 319–334. [DOI] [PubMed] [Google Scholar]
- 3.Neoptolemos JP, Kleeff J, Michl P, Costello E, Greenhalf W, and Palmer DH 2018. Therapeutic developments in pancreatic cancer: current and future perspectives. Nat Rev Gastroenterol Hepatol 15: 333–348. [DOI] [PubMed] [Google Scholar]
- 4.Gleeson FC, and Levy MJ 2018. Genomic Profiling and Potentially Targetable Alterations in Pancreatic Ductal Adenocarcinoma. Curr Treat Options Gastroenterol [DOI] [PubMed]
- 5.Goodwin GH, Sanders C, and Johns EW 1973. A new group of chromatin-associated proteins with a high content of acidic and basic amino acids. Eur J Biochem 38: 14–19. [DOI] [PubMed] [Google Scholar]
- 6.Bustin M 2001. Revised nomenclature for high mobility group (HMG) chromosomal proteins. Trends Biochem Sci 26: 152–153. [DOI] [PubMed] [Google Scholar]
- 7.Kang R, Zhang Q, Zeh HJ 3rd, Lotze MT, and Tang D 2013. HMGB1 in cancer: good, bad, or both? Clin Cancer Res 19: 4046–4057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Tang D, Kang R, Zeh HJ 3rd, and Lotze MT 2010. High-mobility group box 1 and cancer. Biochim Biophys Acta 1799: 131–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sharman AC, Hay-Schmidt A, and Holland PW 1997. Cloning and analysis of an HMG gene from the lamprey Lampetra fluviatilis: gene duplication in vertebrate evolution. Gene 184: 99–105. [DOI] [PubMed] [Google Scholar]
- 10.Celona B, Weiner A, Di Felice F, Mancuso FM, Cesarini E, Rossi RL, Gregory L, Baban D, Rossetti G, Grianti P, Pagani M, Bonaldi T, Ragoussis J, Friedman N, Camilloni G, Bianchi ME, and Agresti A 2011. Substantial histone reduction modulates genomewide nucleosomal occupancy and global transcriptional output. PLoS Biol 9: e1001086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Yu SS, Li HJ, Goodwin GH, and Johns EW 1977. Interaction of non-histone chromosomal proteins HMG1 and HMG2 with DNA. Eur J Biochem 78: 497–502. [DOI] [PubMed] [Google Scholar]
- 12.Jain A, Akanchha S, and Rajeswari MR 2005. Stabilization of purine motif DNA triplex by a tetrapeptide from the binding domain of HMGBI protein. Biochimie 87: 781–790. [DOI] [PubMed] [Google Scholar]
- 13.Gaillard C, and Strauss F 1994. Association of poly(CA).poly(TG) DNA fragments into four-stranded complexes bound by HMG1 and 2. Science 264: 433–436. [DOI] [PubMed] [Google Scholar]
- 14.Hill DA, and Reeves R 1997. Competition between HMG-I(Y), HMG-1 and histone H1 on four-way junction DNA. Nucleic Acids Res 25: 3523–3531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Teo SH, Grasser KD, Hardman CH, Broadhurst RW, Laue ED, and Thomas JO 1995. Two mutations in the HMG-box with very different structural consequences provide insights into the nature of binding to four-way junction DNA. Embo J 14: 3844–3853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Pasheva EA, Pashev IG, and Favre A 1998. Preferential binding of high mobility group 1 protein to UV-damaged DNA. Role of the COOH-terminal domain. J Biol Chem 273: 24730–24736. [DOI] [PubMed] [Google Scholar]
- 17.Javaherian K, Liu JF, and Wang JC 1978. Nonhistone proteins HMG1 and HMG2 change the DNA helical structure. Science 199: 1345–1346. [DOI] [PubMed] [Google Scholar]
- 18.Paull TT, Haykinson MJ, and Johnson RC 1993. The nonspecific DNA-binding and -bending proteins HMG1 and HMG2 promote the assembly of complex nucleoprotein structures. Genes Dev 7: 1521–1534. [DOI] [PubMed] [Google Scholar]
- 19.Lange SS, and Vasquez KM 2009. HMGB1: the jack-of-all-trades protein is a master DNA repair mechanic. Mol Carcinog 48: 571–580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Tang D, Billiar TR, and Lotze MT 2012. A Janus tale of two active high mobility group box 1 (HMGB1) redox states. Mol Med 18: 1360–1362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Tang D, Kang R, Livesey KM, Cheh CW, Farkas A, Loughran P, Hoppe G, Bianchi ME, Tracey KJ, Zeh HJ 3rd, and Lotze MT 2010. Endogenous HMGB1 regulates autophagy. J Cell Biol 190: 881–892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Lee H, Shin N, Song M, Kang UB, Yeom J, Lee C, Ahn YH, Yoo JS, Paik YK, and Kim H 2010. Analysis of nuclear high mobility group box 1 (HMGB1)-binding proteins in colon cancer cells: clustering with proteins involved in secretion and extranuclear function. J Proteome Res 9: 4661–4670. [DOI] [PubMed] [Google Scholar]
- 23.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, and Tracey KJ 1999. HMG-1 as a late mediator of endotoxin lethality in mice. Science 285: 248–251. [DOI] [PubMed] [Google Scholar]
- 24.Tsung A, Sahai R, Tanaka H, Nakao A, Fink MP, Lotze MT, Yang H, Li J, Tracey KJ, Geller DA, and Billiar TR 2005. The nuclear factor HMGB1 mediates hepatic injury after murine liver ischemia-reperfusion. J Exp Med 201: 1135–1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, Huang J, Yu Y, Fan XG, Yan Z, Sun X, Wang H, Wang Q, Tsung A, Billiar TR, Zeh HJ 3rd, Lotze MT, and Tang D 2014. HMGB1 in health and disease. Mol Aspects Med 40: 1–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yu Y, Tang D, and Kang R 2015. Oxidative stress-mediated HMGB1 biology. Front Physiol 6: 93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.He Q, You H, Li XM, Liu TH, Wang P, and Wang BE 2012. HMGB1 Promotes the Synthesis of Pro-IL-1beta and Pro-IL-18 by Activation of p38 MAPK and NF-kappaB Through Receptors for Advanced Glycation End-products in Macrophages. Asian Pac J Cancer Prev 13: 1365–1370. [DOI] [PubMed] [Google Scholar]
- 28.Andersson U, Wang H, Palmblad K, Aveberger AC, Bloom O, Erlandsson-Harris H, Janson A, Kokkola R, Zhang M, Yang H, and Tracey KJ 2000. High mobility group 1 protein (HMG-1) stimulates proinflammatory cytokine synthesis in human monocytes. J Exp Med 192: 565–570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Park JS, Arcaroli J, Yum HK, Yang H, Wang H, Yang KY, Choe KH, Strassheim D, Pitts TM, Tracey KJ, and Abraham E 2003. Activation of gene expression in human neutrophils by high mobility group box 1 protein. Am J Physiol Cell Physiol 284: C870–879. [DOI] [PubMed] [Google Scholar]
- 30.Silva E, Arcaroli J, He Q, Svetkauskaite D, Coldren C, Nick JA, Poch K, Park JS, Banerjee A, and Abraham E 2007. HMGB1 and LPS induce distinct patterns of gene expression and activation in neutrophils from patients with sepsis-induced acute lung injury. Intensive Care Med 33: 1829–1839. [DOI] [PubMed] [Google Scholar]
- 31.Pedrazzi M, Patrone M, Passalacqua M, Ranzato E, Colamassaro D, Sparatore B, Pontremoli S, and Melloni E 2007. Selective proinflammatory activation of astrocytes by high-mobility group box 1 protein signaling. J Immunol 179: 8525–8532. [DOI] [PubMed] [Google Scholar]
- 32.Hreggvidsdottir HS, Ostberg T, Wahamaa H, Schierbeck H, Aveberger AC, Klevenvall L, Palmblad K, Ottosson L, Andersson U, and Harris HE 2009. The alarmin HMGB1 acts in synergy with endogenous and exogenous danger signals to promote inflammation. J Leukoc Biol 86: 655–662. [DOI] [PubMed] [Google Scholar]
- 33.Hou CH, Fong YC, and Tang CH 2011. HMGB-1 induces IL-6 production in human synovial fibroblasts through c-Src, Akt and NF-kappaB pathways. J Cell Physiol 226: 2006–2015. [DOI] [PubMed] [Google Scholar]
- 34.Guo HF, Liu SX, Zhang YJ, Liu QJ, Hao J, and Gao LX 2011. High mobility group box 1 induces synoviocyte proliferation in rheumatoid arthritis by activating the signal transducer and activator transcription signal pathway. Clin Exp Med 11: 65–74. [DOI] [PubMed] [Google Scholar]
- 35.Dejean E, Foisseau M, Lagarrigue F, Lamant L, Prade N, Marfak A, Delsol G, Giuriato S, Gaits-Iacovoni F, and Meggetto F 2012. ALK+ALCLs induce cutaneous, HMGB-1-dependent IL-8/CXCL8 production by keratinocytes through NF-kappaB activation. Blood 119: 4698–4707. [DOI] [PubMed] [Google Scholar]
- 36.Yang D, Chen Q, Yang H, Tracey KJ, Bustin M, and Oppenheim JJ 2007. High mobility group box-1 protein induces the migration and activation of human dendritic cells and acts as an alarmin. J Leukoc Biol 81: 59–66. [DOI] [PubMed] [Google Scholar]
- 37.Bianchi ME 2009. HMGB1 loves company. J Leukoc Biol 86: 573–576. [DOI] [PubMed] [Google Scholar]
- 38.Whitcomb DC 2006. Clinical practice. Acute pancreatitis. N Engl J Med 354: 2142–2150. [DOI] [PubMed] [Google Scholar]
- 39.Shah AU, Sarwar A, Orabi AI, Gautam S, Grant WM, Park AJ, Liu J, Mistry PK, Jain D, and Husain SZ 2009. Protease activation during in vivo pancreatitis is dependent on calcineurin activation. Am J Physiol Gastrointest Liver Physiol 297: G967–973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Satoh A, Shimosegawa T, Kimura K, Moriizumi S, Masamune A, Koizumi M, and Toyota T 1998. Nitric oxide is overproduced by peritoneal macrophages in rat taurocholate pancreatitis: the mechanism of inducible nitric oxide synthase expression. Pancreas 17: 402–411. [DOI] [PubMed] [Google Scholar]
- 41.Sugita H, Yamaguchi Y, Ikei S, Yamada S, and Ogawa M 1997. Enhanced expression of cytokine-induced neutrophil chemoattractant (CINC) by bronchoalveolar macrophages in cerulein-induced pancreatitis rats. Dig Dis Sci 42: 154–160. [DOI] [PubMed] [Google Scholar]
- 42.Gea-Sorli S, and Closa D 2010. Role of macrophages in the progression of acute pancreatitis. World journal of gastrointestinal pharmacology and therapeutics 1: 107–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Shrivastava P, and Bhatia M 2010. Essential role of monocytes and macrophages in the progression of acute pancreatitis. World J Gastroenterol 16: 3995–4002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Hegyi P, Pandol S, Venglovecz V, and Rakonczay Z Jr. 2011. The acinar-ductal tango in the pathogenesis of acute pancreatitis. Gut 60: 544–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Yasuda T, Ueda T, Takeyama Y, Shinzeki M, Sawa H, Nakajima T, Ajiki T, Fujino Y, Suzuki Y, and Kuroda Y 2006. Significant increase of serum high-mobility group box chromosomal protein 1 levels in patients with severe acute pancreatitis. Pancreas 33: 359–363. [DOI] [PubMed] [Google Scholar]
- 46.Yang ZY, Ling Y, Yin T, Tao J, Xiong JX, Wu HS, and Wang CY 2008. Delayed ethyl pyruvate therapy attenuates experimental severe acute pancreatitis via reduced serum high mobility group box 1 levels in rats. World J Gastroenterol 14: 4546–4550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Yuan H, Jin X, Sun J, Li F, Feng Q, Zhang C, Cao Y, and Wang Y 2009. Protective effect of HMGB1 a box on organ injury of acute pancreatitis in mice. Pancreas 38: 143–148. [DOI] [PubMed] [Google Scholar]
- 48.Sawa H, Ueda T, Takeyama Y, Yasuda T, Shinzeki M, Nakajima T, and Kuroda Y 2006. Blockade of high mobility group box-1 protein attenuates experimental severe acute pancreatitis. World J Gastroenterol 12: 7666–7670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zhang ZW, Zhang QY, Zhou MT, Liu NX, Chen TK, Zhu YF, and Wu L 2010. Antioxidant inhibits HMGB1 expression and reduces pancreas injury in rats with severe acute pancreatitis. Dig Dis Sci 55: 2529–2536. [DOI] [PubMed] [Google Scholar]
- 50.Hagiwara S, Iwasaka H, Shingu C, Matsumoto S, Uchida T, and Noguchi T 2009. Antithrombin III prevents cerulein-induced acute pancreatitis in rats. Pancreas 38: 746–751. [DOI] [PubMed] [Google Scholar]
- 51.Hagiwara S, Iwasaka H, Uchida T, Hasegawa A, Asai N, and Noguchi T 2009. Danaparoid sodium prevents cerulein-induced acute pancreatitis in rats. Shock 32: 94–99. [DOI] [PubMed] [Google Scholar]
- 52.Kang R, Zhang Q, Hou W, Yan Z, Chen R, Bonaroti J, Bansal P, Billiar TR, Tsung A, Wang Q, Bartlett DL, Whitcomb DC, Chang EB, Zhu X, Wang H, Lu B, Tracey KJ, Cao L, Fan XG, Lotze MT, Zeh HJ 3rd, and Tang D 2014. Intracellular Hmgb1 inhibits inflammatory nucleosome release and limits acute pancreatitis in mice. Gastroenterology 146: 1097–1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Cox AD, Fesik SW, Kimmelman AC, Luo J, and Der CJ 2014. Drugging the undruggable RAS: Mission possible? Nature reviews. Drug discovery 13: 828–851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Stephen AG, Esposito D, Bagni RK, and McCormick F 2014. Dragging ras back in the ring. Cancer cell 25: 272–281. [DOI] [PubMed] [Google Scholar]
- 55.Hruban RH, and Adsay NV 2009. Molecular classification of neoplasms of the pancreas. Hum Pathol 40: 612–623. [DOI] [PubMed] [Google Scholar]
- 56.Hingorani SR, Petricoin EF, Maitra A, Rajapakse V, King C, Jacobetz MA, Ross S, Conrads TP, Veenstra TD, Hitt BA, Kawaguchi Y, Johann D, Liotta LA, Crawford HC, Putt ME, Jacks T, Wright CV, Hruban RH, Lowy AM, and Tuveson DA 2003. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell 4: 437–450. [DOI] [PubMed] [Google Scholar]
- 57.Kang R, Xie Y, Zhang Q, Hou W, Jiang Q, Zhu S, Liu J, Zeng D, Wang H, Bartlett DL, Billiar TR, Zeh HJ 3rd, Lotze MT, and Tang D 2017. Intracellular HMGB1 as a novel tumor suppressor of pancreatic cancer. Cell Res 27: 916–932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Magruder JT, Elahi D, and Andersen DK 2011. Diabetes and pancreatic cancer: chicken or egg? Pancreas 40: 339–351. [DOI] [PubMed] [Google Scholar]
- 59.Stevens RJ, Roddam AW, and Beral V 2007. Pancreatic cancer in type 1 and young-onset diabetes: systematic review and meta-analysis. Br J Cancer 96: 507–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Everhart J, and Wright D 1995. Diabetes mellitus as a risk factor for pancreatic cancer. A meta-analysis. JAMA 273: 1605–1609. [PubMed] [Google Scholar]
- 61.Gullo L, Pezzilli R, and Morselli-Labate AM 1994. Diabetes and the risk of pancreatic cancer. N Engl J Med 331: 81–84. [DOI] [PubMed] [Google Scholar]
- 62.Muniraj T, and Chari ST 2012. Diabetes and pancreatic cancer. Minerva Gastroenterol Dietol 58: 331–345. [PMC free article] [PubMed] [Google Scholar]
- 63.Biscetti F, Straface G, De Cristofaro R, Lancellotti S, Rizzo P, Arena V, Stigliano E, Pecorini G, Egashira K, De Angelis G, Ghirlanda G, and Flex A 2010. High-mobility group box-1 protein promotes angiogenesis after peripheral ischemia in diabetic mice through a VEGF-dependent mechanism. Diabetes 59: 1496–1505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Yang Z, and Klionsky DJ 2010. Eaten alive: a history of macroautophagy. Nat Cell Biol 12: 814–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Klionsky DJ, and Emr SD 2000. Autophagy as a regulated pathway of cellular degradation. Science 290: 1717–1721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.White E 2012. Deconvoluting the context-dependent role for autophagy in cancer. Nat Rev Cancer 12: 401–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Rosenfeldt MT, O’Prey J, Morton JP, Nixon C, Mackay G, Mrowinska A, Au A, Rai TS, Zheng L, Ridgway R, Adams PD, Anderson KI, Gottlieb E, Sansom OJ, and Ryan KM 2013. p53 status determines the role of autophagy in pancreatic tumour development. Nature. [DOI] [PubMed]
- 68.Li C, Zhang Y, Cheng X, Yuan H, Zhu S, Liu J, Wen Q, Xie Y, Liu J, Kroemer G, Klionsky DJ, Lotze MT, Zeh HJ, Kang R, and Tang D 2018. PINK1 and PARK2 Suppress Pancreatic Tumorigenesis through Control of Mitochondrial Iron-Mediated Immunometabolism. Dev Cell 46: 441–455 e448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Yang S, Wang X, Contino G, Liesa M, Sahin E, Ying H, Bause A, Li Y, Stommel JM, Dell’antonio G, Mautner J, Tonon G, Haigis M, Shirihai OS, Doglioni C, Bardeesy N, and Kimmelman AC 2011. Pancreatic cancers require autophagy for tumor growth. Genes Dev 25: 717–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Perera RM, Stoykova S, Nicolay BN, Ross KN, Fitamant J, Boukhali M, Lengrand J, Deshpande V, Selig MK, Ferrone CR, Settleman J, Stephanopoulos G, Dyson NJ, Zoncu R, Ramaswamy S, Haas W, and Bardeesy N 2015. Transcriptional control of autophagy-lysosome function drives pancreatic cancer metabolism. Nature 524: 361–365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Gonzalez CD, Lee MS, Marchetti P, Pietropaolo M, Towns R, Vaccaro MI, Watada H, and Wiley JW 2011. The emerging role of autophagy in the pathophysiology of diabetes mellitus. Autophagy 7: 2–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Jung HS, Chung KW, Won Kim J, Kim J, Komatsu M, Tanaka K, Nguyen YH, Kang TM, Yoon KH, Kim JW, Jeong YT, Han MS, Lee MK, Kim KW, Shin J, and Lee MS 2008. Loss of autophagy diminishes pancreatic beta cell mass and function with resultant hyperglycemia. Cell Metab 8: 318–324. [DOI] [PubMed] [Google Scholar]
- 73.Ebato C, Uchida T, Arakawa M, Komatsu M, Ueno T, Komiya K, Azuma K, Hirose T, Tanaka K, Kominami E, Kawamori R, Fujitani Y, and Watada H 2008. Autophagy is important in islet homeostasis and compensatory increase of beta cell mass in response to high-fat diet. Cell Metab 8: 325–332. [DOI] [PubMed] [Google Scholar]
- 74.Quan W, Lim YM, and Lee MS 2012. Role of autophagy in diabetes and endoplasmic reticulum stress of pancreatic beta-cells. Exp Mol Med 44: 81–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Tang D, Kang R, Livesey KM, Kroemer G, Billiar TR, Van Houten B, Zeh HJ 3rd, and Lotze MT 2011. High-mobility group box 1 is essential for mitochondrial quality control. Cell Metabolism 13: 701–711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Livesey KM, Kang R, Vernon P, Buchser W, Loughran P, Watkins SC, Zhang L, Manfredi JJ, Zeh HJ 3rd, Li L, Lotze MT, and Tang D 2012. p53/HMGB1 complexes regulate autophagy and apoptosis. Cancer Res 72: 1996–2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Huang J, Ni J, Liu K, Yu Y, Xie M, Kang R, Vernon P, Cao L, and Tang D 2012. HMGB1 promotes drug resistance in osteosarcoma. Cancer Res 72: 230–238. [DOI] [PubMed] [Google Scholar]
- 78.Tang D, Kang R, Cheh CW, Livesey KM, Liang X, Schapiro NE, Benschop R, Sparvero LJ, Amoscato AA, Tracey KJ, Zeh HJ, and Lotze MT 2010. HMGB1 release and redox regulates autophagy and apoptosis in cancer cells. Oncogene 29: 5299–5310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Huebener P, Gwak GY, Pradere JP, Quinzii CM, Friedman R, Lin CS, Trent CM, Mederacke I, Zhao E, Dapito DH, Lin Y, Goldberg IJ, Czaja MJ, and Schwabe RF 2014. High-mobility group box 1 is dispensable for autophagy, mitochondrial quality control, and organ function in vivo. Cell Metab 19: 539–547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Sun X, and Tang D 2014. HMGB1-dependent and -independent autophagy. Autophagy 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Sims GP, Rowe DC, Rietdijk ST, Herbst R, and Coyle AJ 2010. HMGB1 and RAGE in inflammation and cancer. Annual review of immunology 28: 367–388. [DOI] [PubMed] [Google Scholar]
- 82.Sparvero LJ, Asafu-Adjei D, Kang R, Tang D, Amin N, Im J, Rutledge R, Lin B, Amoscato AA, Zeh HJ, and Lotze MT 2009. RAGE (Receptor for Advanced Glycation Endproducts), RAGE ligands, and their role in cancer and inflammation. Journal of translational medicine 7: 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Gebhardt C, Riehl A, Durchdewald M, Nemeth J, Furstenberger G, Muller-Decker K, Enk A, Arnold B, Bierhaus A, Nawroth PP, Hess J, and Angel P 2008. RAGE signaling sustains inflammation and promotes tumor development. J Exp Med 205: 275–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Taguchi A, Blood DC, del Toro G, 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, and Schmidt AM 2000. Blockade of RAGE-amphoterin signalling suppresses tumour growth and metastases. Nature 405: 354–360. [DOI] [PubMed] [Google Scholar]
- 85.Akirav EM, Preston-Hurlburt P, Garyu J, Henegariu O, Clynes R, Schmidt AM, and Herold KC 2012. RAGE expression in human T cells: a link between environmental factors and adaptive immune responses. PLoS ONE 7: e34698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Chen X, Zhang L, Zhang IY, Liang J, Wang H, Ouyang M, Wu S, da Fonseca AC, Weng L, Yamamoto Y, Yamamoto H, Natarajan R, and Badie B 2014. RAGE expression in tumor-associated macrophages promotes angiogenesis in glioma. Cancer Res 74: 7285–7297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Xiong F, Leonov S, Howard AC, Xiong S, Zhang B, Mei L, McNeil P, Simon S, and Xiong WC 2011. Receptor for advanced glycation end products (RAGE) prevents endothelial cell membrane resealing and regulates F-actin remodeling in a beta-catenin-dependent manner. J Biol Chem 286: 35061–35070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Hou FF, Jiang JP, Guo JQ, Wang GB, Zhang X, Stern DM, Schmidt AM, and Owen WF Jr. 2002. Receptor for advanced glycation end products on human synovial fibroblasts: role in the pathogenesis of dialysis-related amyloidosis. J Am Soc Nephrol 13: 1296–1306. [DOI] [PubMed] [Google Scholar]
- 89.Vernon PJ, Loux TJ, Schapiro NE, Kang R, Muthuswamy R, Kalinski P, Tang D, Lotze MT, and Zeh HJ 3rd. 2013. The receptor for advanced glycation end products promotes pancreatic carcinogenesis and accumulation of myeloid-derived suppressor cells. J Immunol 190: 1372–1379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Kang R, Loux T, Tang D, Schapiro NE, Vernon P, Livesey KM, Krasinskas A, Lotze MT, and Zeh HJ 3rd. 2012. The expression of the receptor for advanced glycation endproducts (RAGE) is permissive for early pancreatic neoplasia. Proceedings of the National Academy of Sciences of the United States of America 109: 7031–7036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Kang R, Tang D, Livesey KM, Schapiro NE, Lotze MT, and Zeh HJ 3rd. 2011. The Receptor for Advanced Glycation End-products (RAGE) protects pancreatic tumor cells against oxidative injury. Antioxid Redox Signal 15: 2175–2184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Kang R, Tang D, Schapiro NE, Livesey KM, Farkas A, Loughran P, Bierhaus A, Lotze MT, and Zeh HJ 2010. The receptor for advanced glycation end products (RAGE) sustains autophagy and limits apoptosis, promoting pancreatic tumor cell survival. Cell death and differentiation 17: 666–676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Kang R, Tang D, Schapiro NE, Loux T, Livesey KM, Billiar TR, Wang H, Van Houten B, Lotze MT, and Zeh HJ 2014. The HMGB1/RAGE inflammatory pathway promotes pancreatic tumor growth by regulating mitochondrial bioenergetics. Oncogene 33: 567–577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kang R, Hou W, Zhang Q, Chen R, Lee YJ, Bartlett DL, Lotze MT, Tang D, and Zeh HJ 2014. RAGE is essential for oncogenic KRAS-mediated hypoxic signaling in pancreatic cancer. Cell Death Dis 5: e1480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kang R, Tang D, Lotze MT, and Zeh HJ 3rd. 2012. AGER/RAGE-mediated autophagy promotes pancreatic tumorigenesis and bioenergetics through the IL6-pSTAT3 pathway. Autophagy 8: 989–991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kang R, Tang D, Lotze MT, and Zeh HJ 3rd. 2011. RAGE regulates autophagy and apoptosis following oxidative injury. Autophagy 7: 442–444. [DOI] [PubMed] [Google Scholar]
- 97.Kang R, Tang D, Loze MT, and Zeh HJ 2011. Apoptosis to autophagy switch triggered by the MHC class III-encoded receptor for advanced glycation endproducts (RAGE). Autophagy 7: 91–93. [DOI] [PubMed] [Google Scholar]
- 98.Sirois CM, Jin T, Miller AL, Bertheloot D, Nakamura H, Horvath GL, Mian A, Jiang J, Schrum J, Bossaller L, Pelka K, Garbi N, Brewah Y, Tian J, Chang C, Chowdhury PS, Sims GP, Kolbeck R, Coyle AJ, Humbles AA, Xiao TS, and Latz E 2013. RAGE is a nucleic acid receptor that promotes inflammatory responses to DNA. The Journal of experimental medicine 210: 2447–2463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Kang R, Chen R, Xie M, Cao L, Lotze MT, Tang D, and Zeh HJ 3rd. 2016. The Receptor for Advanced Glycation End Products Activates the AIM2 Inflammasome in Acute Pancreatitis. J Immunol 196: 4331–4337. [DOI] [PMC free article] [PubMed] [Google Scholar]