High mobility group box 1 and kidney diseases (Review)

  • Authors:
    • Ping Zhu
    • Li Xie
    • Hua-Sheng Ding
    • Quan Gong
    • Jian Yang
    • Lin Yang
  • View Affiliations

  • Published online on: February 25, 2013     https://doi.org/10.3892/ijmm.2013.1286
  • Pages: 763-768
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

High mobility group box 1 (HMGB1), a non-histone DNA-binding protein, regulates nucleosome function and transcription in the nuclei of all metazoans and plants. However, extracellular HMGB1, which is actively or passively released under different conditions, can act as a key inflammatory mediator through MyD88/mitogen-activated protein kinase signaling by binding to its receptors including the receptor for advanced glycation end products or Toll-like receptors. A growing body of evidence indicates that HMGB1 plays an important role in kidney diseases, such as glomerulonephritis, lupus nephritis, antineutrophilic cytoplasmatic antibody-associated vaculitis, diabetic nephropathy, renal allograft rejection and acute kidney injury. In this review, we focus on the biology of HMGB1 and the association of HMGB1 with kidney diseases.

1. Introduction

High mobility group box 1 (HMGB1) was originally described 30 years ago as a nonhistone DNA-binding protein with high-electrophoretic mobility (1). It is present in the nucleus of almost all metazoans and plants, where it exerts structural and transcriptional activities (24). In addition to its role in the nucleus, HMGB1 has recently emerged as an extracellular signaling factor with key roles in cell differentiation, proliferation and disease pathogenesis (5).

In this review, we focus on the involvement of HMGB1 in the pathogenesis of kidney diseases, as well as renal allograft rejection after renal transplantation. An understanding of these discoveries may shed new insight into the possibilities for developing novel therapeutic strategies to mitigate or prevent kidney diseases.

2. Structure of HMGB1

The HMGB1 protein, a member of the high mobility group nuclear protein family, is one of the most evolutionarily conserved proteins and shares 99% identity in the amino acid sequence between rodents and humans (1). The human HMGB1 gene is located on chromosome 13q12 (6), and six polymorphic loci throughout the gene locus have recently been identified (7).

HMGB1 has a highly dipolar structure consisting of 215 residues organized into two basic DNA-binding domains, referred to as the A and B box as well as a negatively charged C-acidic tail (Fig. 1) (8). Each HMGB1 A or B box is approximately 75–80 amino acids in length (9) and is formed by two short and one long α-helix that upon folding produce an L- or V-shaped three-dimensional domain structure (10,11). Research suggests that the B box possesses the pro-inflammatory properties of HMGB1 including cytokine release, and the A box instead competes with HMGB1 for binding sites leading to attenuation of the inflammatory cascade (12,13).

3. Function of intra-nuclear HMGB1

As the HMGB1 protein is essential for life, HMGB1-knockout mice die shortly after birth (14). HMGB1 is fairly ubiquitous in mammals and is almost always present in the nucleus (15)where HMGB1 plays an important role in binding without sequence specificity to the minor groove of DNA and induces bends in the helical structure, which regulate physical interactions between DNA and transcription factors, including p53, homeobox proteins, glucocorticoid receptor, recombination-activating gene 1/2 (RAG1/2) proteins and steroid hormone receptors (16,17). Even though HMGB1 is not essential for the overall organization of chromatin in the nucleus, it is critical for proper transcriptional control by specific transcription factors (14,18).

4. Release of HMGB1

In addition to its roles in regulating nucleosome function and transcription, HMGB1 has recently emerged as an extracellularly release mediator of inflammation, although it lacks a classical secretion signal. There are two modes of HMGB1 release: one active and one passive. HMGB1 not only is passively released from damaged cells under conditions of injury, but can also be actively secreted from activated immune cells in response to inflammatory stimuli (18,19).

How is HMGB1 released? A growing body of evidence indicates that inflammasomes play an important role in the release of HMGB1. Recently, NLRP3 inflammasome activation was shown to be essential for HMGB1 release from LPS-primed macrophages treated with ATP or exposed to nigericin (20). Similarly, the extracellular release of HMGB1 from S. typhimurium-infected macrophages was found to rely on activation of caspase-1 by the NLRC4 inflammasome (20). However, the exact mechanism remains unclear (Fig. 2).

5. Ligands and signaling of HMGB1

To date, several important surface receptors have been implicated in HMGB1 signaling, including the receptor for advanced glycation end products (RAGE), Toll-like receptor (TLR)2, TLR4 and syndecan (21,22). Binding to these receptors results in the activation of nuclear factor (NF)-κB, which induces the upregulation of pro-inflammatory cytokines, thereby promoting inflammation.

RAGE which is expressed on monocytes, macrophages, neurons and endothelial cells, as well as on a variety of tumor cells is thought to be one of the primary receptors for HMGB1 (23,24). Interaction of HMGB1 with RAGE can activate two major signaling pathways, one encompassing CDC42/Rac and the other involving diverse mitogen-activated protein kinases (MAPKs) that finally leads to cytoskeletal changes and NF-κB activation, respectively (25,26).

In addition to RAGE, TLR2 and 4 which are expressed on antigen-presenting cells (APCs) were found to directly interact with HMGB1 as determined by fluorescence resonance energy transfer (FRET) and immunoprecipitation (27). Numerous studies have demonstrated that HMGB1 signaling through TLR2 and TLR4 is mediated by the Rac1/phosphoinositide-3-kinase (PI3K)/CDC42 pathway and MyD88-dependent NF-κB activation pathway, respectively (21,28,29).

Recently, numerous studies have 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 (30) which is considered as an accelerator of TLR9 response with CpG-DNA (31). HMGB1 accelerates the delivery of CpG-ODNs to its receptor, leading to a TLR9-dependent augmentation of interleukin (IL)-6, IL-12 and tumor necrosis factor (TNF)-α secretion (30).

More recently, apart from RAGE and TLRs, HMGB1 can interact with a wide range of proteins with a phage display approach (10). Yet, further studies are required to determine the exact function of HMGB1 interaction with these proteins.

6. HMGB1 and kidney diseases

HMGB1 and glomerulonephritis

Granulomatous nephritis is triggered by a diverse group of factors and results in renal failure. Granulomatous inflammation is one of the most significant pathogenetic mechanisms in nephritis. Recently, research revealed that the HMGB1 level in urine and serum was elevated in crystal-induced granulomatous nephritis caused by an adenine-rich diet, and HMGB1 induced monocyte chemoattractant protein-1 (MCP-1) secretion through the MAPK and PI3K pathways. The authors concluded that HMGB1 is a new mediator involved in crystal-induced nephritis that amplifies granulomatous inflammation through a cycle in which MCP-1 attracts activated macrophages, resulting in excessive and sustained HMGB1 release. Thus, HMGB1 may be a novel target for the prevention or treatment of granulomatous nephritis (32).

HMGB1 and secondary kidney diseases
HMGB1 and lupus nephritis

Lupus nephritis is common in systemic lupus erythematosus (SLE) patients and manifests mainly by proteinuria, hematuria, and, less commonly, severe renal failure. The mechanism of lupus nephritis is not fully clear. Yet, accumulating data suggest that HMGB1 may play an important role in lupus nephritis.

Iwata et al (33) revealed that HMGB1 secreted by dendritic cells via p38 MAPK activation participates in autoimmunity in MRL-Fas(lpr) mice (a lupus-prone mouse model). These results suggest that HMGB1 is involved in the progression of autoimmune kidney diseases in MRL-Fas(lpr) mice (33).

Research reveals that high expression of HMGB1 (in blood or renal biopsies) is positively correlated with MCP-1 expression and may contribute to the pathogenesis of lupus nephritis (34,35). Recently, Feng et al (36) demonstrated that HMGB1 mRNA and protein levels were increased in the glomeruli of lupus nephritis patients and BXSB mice. Their findings indicate that HMGB1 mediates interferon (IFN)-γ-induced cell proliferation in mouse mesangial cells through regulation of the cyclin D1/CDK4/p16 pathway and promotion of cell cycle transition from G1 to S stage in lupus nephritis (36). Therefore, the above findings clearly indicate a key role of HMGB1 in lupus nephritis, and inhibition of HMGB1 or MCP-1 may be a novel treatment strategy for lupus nephritis (34,37).

HMGB1 and antineutrophilic cytoplasmatic antibody (ACNA)-associated vasculitis (AAV)

Cytokines, such as TNF and IL-6, are important in ACNA-AAV. HMGB1 can induce the release of TNF and IL-6 by APCs. Recently, Bruchfeld et al (38) concluded that the level of HMGB1 is increased in AVV with renal manifestations. This suggests that HMGB1 plays an important role in AVV.

HMGB1 and diabetic nephropathy

Research has shown that the expression of HSP70 and HMGB1, endogenous ligands of TLRs, is significantly upregulated in the kidneys of diabetic rats. These findings suggest that release of hyperglycemia-induced HMGB1 may induce renal injury in diabetic rats, and that the pathogenic role of HMGB1 may be dependent on RAGE or TLR4 and through activation of NF-κB and may promote tubulointerstitial inflammation in diabetic nephropathy (39,40). In contrast, other results found that the level of HMGB1 in serum was decreased in patients with diabetic nephropathy (41). Further extensive study is required to explore the role of HMGB1 in diabetic nephropathy.

HMGB1 and autosomal dominant polycystic kidney disease

Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenic kidney disease and the fourth leading cause of end-stage kidney disease in adults worldwide (42). Recently, two research groups reported that the serum level of HMGB1 was increased in ADPKD patients (43,44) which suggests that HMGB1 may play a role in ADPKD.

HMGB1 and acute kidney injury

In 1999, HMGB1 was implicated as a late mediator of lethal systemic inflammation in sepsis (18). As acute kidney injury is a severe complication of sepsis, there is evidence to indicate that HMGB1 plays a significant role in sepsis-mediated acute kidney injury (45).

Wang et al (46) found that HMGB1 expression was markedly increased in renal tissue and in acute kidney injury in rats with delayed resuscitation after thermal injury. Chen et al (47) found that HMGB1 released by injured renal cells in renal ischemia and reperfusion injury (RIRI) can induce TLR4 (+/+) leukocytes producing IL-6 by binding to its receptor TLR4. This underscores the importance of HMGB1/TLR4 signaling in the pathogenesis of ischemic acute kidney injury.

In addition, Chung et al (48) found that HMGB1 expression was increased in the kidney 6 h after reperfusion and was decreased gradually 1, 3 and 5 days following reperfusion. Moreover, Wu et al (49) demonstrated that mice treated with anti-HMGB1 antibody had significantly less tubulointerstitial infiltration by neutrophils (day 1) and macrophages (day 5) and markedly reduced apoptosis of tubular epithelial cells. Furthermore, anti-HMGB1 antibody-treated IRI kidneys had significantly lower levels of IL-6, TNF and MCP-1 mRNA, which are downstream of HMGB1. Conversely, administration of rHMGB1 after reperfusion exacerbated kidney IRI in wild-type mice. They conclude that HMGB1 contributes to kidney ischemia reperfusion injury (50). These findings demonstrate that HMGB1 plays an important role in acute kidney injury.

HMGB1 and chronic kidney disease

Chronic kidney disease is a multifactorial disorder occurring in the context of chronic conditions of co-morbidity. Research has indicated that chronic kidney disease is an immune inflammatory condition (51). There is accumulating evidence linking HMGB1 and chronic kidney disease.

Bruchfeld et al (52) revealed that HMGB1 is significantly elevated in patients with chronic kidney disease and correlates with GFR as well as markers of inflammation and malnutrition. Another study found that HMGB1 was expressed in the serum of patients with renal diseases who underwent renal biopsies, particularly patients who suffered from vasculitis including ANCA-GN, Henoch-Schonlein purpura nephritis, and IgAN with glomerular crescents (53). Moreover, Leelahavanichkul et al (54) suggested that HMGB1 is an important common mediator for both chronic kidney disease and sepsis.

Nakamura et al (55) demonstrated that HMGB1 enhances the accumulation of asymmetric dimethylarginine (ADMA) levels, suggesting the active involvement of the AGE/HMGB1-RAGE-ADMA axis in nondiabetic chronic kidney disease patients and that inhibition of HMGB1/RAGE may be a strategy for the treatment of chronic kidney disease (56).

In addition, research suggests that HMGB1 is a key mediator of immune-mediated epithelial-mesenchymal transition (EMT) of proximal tubular epithelial cells and a potentially important signaling molecule in the development of renal fibrosis (57).

HMGB1 and clear cell renal cell carcinoma

Renal cell carcinoma is the most common cancer of the kidney. The main histological subtypes are clear cell (75%), papillary (15%) and chromophobe renal cell carcinoma (5%) (58). Recently, Lin et al (59) demonstrated that HMGB1 promotes the development and progression of clear cell renal cell carcinoma via ERK1/2 activation, which is partially mediated by RAGE. This suggests that HMGB1 is involved in clear cell renal cell carcinoma.

HMGB1 and chronic allograft dysfunction

Chronic allograft dysfunction, a leading cause of chronic allograft failure among kidney transplant recipients, is a multifactorial process associated with progressive interstitial fibrosis and tubular atrophy (60). Recently, Wang et al (61) found that the level of HMGB1/TLR4 was increased in chronic renal transplantation patients. Their findings indicate the MyD88 and TRIF signaling plays an important role in graft-infiltrating mononuclear cells in the pathophysiology of chronic allograft dysfunction (61). These findings suggest that HMGB1 may be an effective target for the prevention and treatment of chronic allograft dysfunction.

7. Conclusion

In conclusion, the robust associations between HMGB1 and kidney diseases have been reviewed in this article (Table I). Although the mechanisms promoting the release of HMGB1 and the signaling pathways it activates require further elucidation, evidence suggests its potential as a therapeutic target/agent in various kidney diseases. Considering its notable role in kidney diseases, a therapeutic approach involving the HMGB1-mediated signaling pathway may constitute a new strategy for the treatment of kidney diseases. Future research may aid in determining the feasibility of such an approach.

Table I

The main role of HMGB1 in kidney diseases.

Table I

The main role of HMGB1 in kidney diseases.

Kidney diseasesRole of HMGB1
HMGB1 and glomerulonephritisPromotes inflammation and induces MCP-1 secretion
HMGB1 and secondary kidney diseases
 HMGB1 and lupus nephritisPromotes inflammation and induces MCP-1 secretion; mediates IFN-γ-induced cell proliferation
 HMGB1 and ACNA-associated vasculitisMediates the immune response
 HMGB1 and diabetic nephropathyActivation of NF-κB; chemotactic; promotes tubulointerstitial inflammation
HMGB1 and dominant polycystic kidney diseasePromotes inflammatory injury
HMGB1 and acute kidney injury
 Acute kidney injury of sepsisMediates inflammation; increases influx of neutrophils
 Acute kidney injury of ischemia and reperfusion injuryActivation of macrophages; induces cytokine release; reduces apoptosis of tubular epithelial cells
HMGB1 and chronic kidney diseasesPromotes immune response; enhances ADMA levels; immune-mediated EMT
HMGB1 and renal cell carcinomaActivation of ERK1/2
HMGB1 and chronic allograft dysfunctionPromotes inflammation through MyD88 and TRIF signaling

[i] HMGB1, high mobility group box 1; MCP-1, monocyte chemoattractant protein-1; ADMA, accumulation of asymmetric dimethylarginine; EMT, epithelial-mesenchymal transition; IFN-γ, interferon; NF-κB, nuclear factor.

References

1 

Goodwin GH, Sanders C and Johns EW: A new group of chromatin-associated proteins with a high content of acidic and basic amino acids. Eur J Biochem. 38:14–19. 1973. View Article : Google Scholar : PubMed/NCBI

2 

Bustin M, Lehn D and Landsman D: Structural features of the HMG chromosomal proteins and their genes. Biochim Biophys Acta. 1049:231–243. 1990. View Article : Google Scholar : PubMed/NCBI

3 

Bonaldi T, Längst G, Strohner R, Becker PB and Bianchi ME: The DNA chaperone HMGB1 facilitates ACF/CHRAC-dependent nucleosome sliding. EMBO J. 21:6865–6873. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Travers AA: Priming the nucleosome: a role for HMGB proteins? EMBO Rep. 4:131–136. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Hock R, Furusawa T, Ueda T and Bustin M: HMG chromosomal proteins in development and disease. Trends Cell Biol. 17:72–79. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Ferrari S, Finelli P, Rocchi M and Bianchi M: The active gene that encodes human high mobility group 1 protein (HMG1) contains introns and maps to chromosome 13. Genomics. 35:367–371. 1996. View Article : Google Scholar : PubMed/NCBI

7 

Kornblit B, Munthe-Fog L, Petersen S, Madsen H, Vindeløv L and Garred P: The genetic variation of the human HMGB1 gene. Tissue Antigens. 70:151–156. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Landsman D and Bustin M: A signature for the HMG-1 box DNA-binding proteins. Bioessays. 15:539–546. 1993. View Article : Google Scholar : PubMed/NCBI

9 

Han J, Zhong J, Wei W, et al: Extracellular high-mobility group box 1 acts as an innate immune mediator to enhance autoimmune progression and diabetes onset in NOD mice. Diabetes. 57:2118–2127. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Dintilhac A and Bernués J: HMGB1 interacts with many apparently unrelated proteins by recognizing short amino acid sequences. J Biol Chem. 277:7021–7028. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Weir H, Kraulis P, Hill C, Raine A, Laue E and Thomas J: Structure of the HMG box motif in the B-domain of HMG1. EMBO J. 12:1311–1319. 1993.PubMed/NCBI

12 

Messmer D, Yang H, Telusma G, et al: High mobility group box protein 1: an endogenous signal for dendritic cell maturation and Th1 polarization. J Immunol. 173:307–313. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Yang H, Ochani M, Li J, et al: Reversing established sepsis with antagonists of endogenous high-mobility group box 1. Proc Natl Acad Sci USA. 101:296–301. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Calogero S, Grassi F, Aguzzi A, et al: The lack of chromosomal protein Hmg1 does not disrupt cell growth but causes lethal hypoglycaemia in newborn mice. Nat Genet. 22:276–280. 1999. View Article : Google Scholar : PubMed/NCBI

15 

Müller S, Scaffidi P, Degryse B, et al: New EMBO member’s review: the double life of HMGB1 chromatin protein: architectural factor and extracellular signal. EMBO J. 20:4337–4340. 2001.

16 

Ulloa L, Batliwalla FM, Andersson U, Gregersen PK and Tracey KJ: High mobility group box chromosomal protein 1 as a nuclear protein, cytokine, and potential therapeutic target in arthritis. Arthritis Rheum. 48:876–881. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Brickman JM, Adam M and Ptashne M: Interactions between an HMG-1 protein and members of the Rel family. Proc Natl Acad Sci USA. 96:10679–10683. 1999. View Article : Google Scholar : PubMed/NCBI

18 

Wang H, Bloom O, Zhang M, et al: HMG-1 as a late mediator of endotoxin lethality in mice. Science. 285:248–251. 1999. View Article : Google Scholar : PubMed/NCBI

19 

Bianchi ME and Manfredi AA: High-mobility group box 1 (HMGB1) protein at the crossroads between innate and adaptive immunity. Immunol Rev. 220:35–46. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Lamkanfi M, Sarkar A, Vande Walle L, et al: Inflammasome-dependent release of the alarmin HMGB1 in endotoxemia. J Immunol. 185:4385–4392. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Park JS, Svetkauskaite D, He Q, et al: Involvement of toll-like receptors 2 and 4 in cellular activation by high mobility group box 1 protein. J Biol Chem. 279:7370–7377. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Kokkola R, Andersson A, Mullins G, et al: RAGE is the major receptor for the proinflammatory activity of HMGB1 in rodent macrophages. Scand J Immunol. 61:1–9. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Hori O, Brett J, Slattery T, et al: The receptor for advanced glycation end products (RAGE) is a cellular binding site for amphoterin. J Biol Chem. 270:25752–25761. 1995. View Article : Google Scholar : PubMed/NCBI

24 

Yang H, Wang H, Czura CJ and Tracey KJ: The cytokine activity of HMGB1. J Leukoc Biol. 78:1–8. 2005. View Article : Google Scholar

25 

Merenmies J, Pihlaskari R, Laitinen J, Wartiovaara J and Rauvala H: 30-kDa heparin-binding protein of brain (amphoterin) involved in neurite outgrowth. Amino acid sequence and localization in the filopodia of the advancing plasma membrane. J Biol Chem. 266:16722–16729. 1991.

26 

Taguchi A, Blood DC, del Toro G, et al: Blockade of RAGE-amphoterin signalling suppresses tumour growth and metastases. Nature. 405:354–360. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Park JS, Gamboni-Robertson F, He Q, et al: High mobility group box 1 protein interacts with multiple Toll-like receptors. Am J Physiol Cell Physiol. 290:C917–C924. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Park JS, Arcaroli J, Yum HK, et al: Activation of gene expression in human neutrophils by high mobility group box 1 protein. Am J Physiol Cell Physiol. 284:C870–C879. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Yu M, Wang H, Ding A, et al: HMGB1 signals through toll-like receptor (TLR) 4 and TLR2. Shock. 26:174–179. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Ivanov S, Dragoi AM, Wang X, et al: A novel role for HMGB1 in TLR9-mediated inflammatory responses to CpG-DNA. Blood. 110:1970–1981. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Tian J, Avalos AM, Mao SY, et al: Toll-like receptor 9-dependent activation by DNA-containing immune complexes is mediated by HMGB1 and RAGE. Nat Immunol. 8:487–496. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Oyama Y, Hashiguchi T, Taniguchi N, et al: High-mobility group box-1 protein promotes granulomatous nephritis in adenine-induced nephropathy. Lab Invest. 90:853–866. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Iwata Y, Furuichi K, Sakai N, et al: Dendritic cells contribute to autoimmune kidney injury in MRL-Faslpr mice. J Rheumatol. 36:306–314. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Zhou JG, Dong JY, Zhang LH and Wang J: Expression of high mobility group box chromosomal protein 1 in mice with lupus nephritis. Zhejiang Da Xue Xue Bao Yi Xue Ban. 40:200–206. 2011.(In Chinese).

35 

Pisetsky DS: HMGB1: a smoking gun in lupus nephritis? Arthritis Res Ther. 14:1122012. View Article : Google Scholar : PubMed/NCBI

36 

Feng X, Hao J, Liu Q, et al: HMGB1 mediates IFN-γ-induced cell proliferation in MMC cells through regulation of cyclin D1/CDK4/p16 pathway. J Cell Biochem. 113:2009–2019. 2012.

37 

Zickert A, Palmblad K, Sundelin B, et al: Renal expression and serum levels of high mobility group box 1 protein in lupus nephritis. Arthritis Res Ther. 14:R362012. View Article : Google Scholar : PubMed/NCBI

38 

Bruchfeld A, Wendt M, Bratt J, et al: High-mobility group box-1 protein (HMGB1) is increased in antineutrophilic cytoplasmatic antibody (ANCA)-associated vasculitis with renal manifestations. Mol Med. 17:29–35. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Kim J, Sohn E, Kim CS, Jo K and Kim JS: The role of high-mobility group box-1 protein in the development of diabetic nephropathy. Am J Nephrol. 33:524–529. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Lin M, Yiu WH, Wu HJ, et al: Toll-like receptor 4 promotes tubular inflammation in diabetic nephropathy. J Am Soc Nephrol. 23:86–102. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Penfold SA, Coughlan MT, Patel SK, et al: Circulating high-molecular-weight RAGE ligands activate pathways implicated in the development of diabetic nephropathy. Kidney Int. 78:287–295. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Torres VE, Harris PC and Pirson Y: Autosomal dominant polycystic kidney disease. Lancet. 369:1287–1301. 2007. View Article : Google Scholar : PubMed/NCBI

43 

Nakamura T, Kawagoe Y, Ueda Y, Yamada S and Koide H: Hemoperfusion treatment in a septic shock patient with autosomal dominant polycystic kidney disease and increased HMGB1 protein levels. Blood Purif. 32:139–142. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Nakamura T, Sato E, Fujiwara N, et al: Changes in urinary albumin excretion, inflammatory and oxidative stress markers in ADPKD patients with hypertension. Am J Med Sci. 343:46–51. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Hu YM, Pai MH, Yeh CL, Hou YC and Yeh SL: Glutamine administration ameliorates sepsis-induced kidney injury by downregulating the high-mobility group box protein-1-mediated pathway in mice. Am J Physiol Renal Physiol. 302:F150–F158. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Wang Q, Yao YM, Wang YB, et al: Effect of ethyl pyruvate on renal high mobility group box-1 protein expression and acute kidney injury in rats with delayed resuscitation after thermal injury. Zhonghua Wai Ke Za Zhi. 45:1210–1213. 2007.(In Chinese).

47 

Chen J, Hartono JR, John R, et al: Early interleukin 6 production by leukocytes during ischemic acute kidney injury is regulated by TLR4. Kidney Int. 80:504–515. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Chung KY, Park JJ and Kim YS: The role of high-mobility group box-1 in renal ischemia and reperfusion injury and the effect of ethyl pyruvate. Transplant Proc. 40:2136–2138. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Wu H, Ma J, Wang P, et al: HMGB1 contributes to kidney ischemia reperfusion injury. J Am Soc Nephrol. 21:1878–1890. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Li J, Gong Q, Zhong S, et al: Neutralization of the extracellular HMGB1 released by ischaemic damaged renal cells protects against renal ischaemia-reperfusion injury. Nephrol Dial Transplant. 26:469–478. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Bao YS, Na SP, Zhang P, et al: Characterization of interleukin-33 and soluble ST2 in serum and their association with disease severity in patients with chronic kidney disease. J Clin Immunol. 32:587–594. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Bruchfeld A, Qureshi AR, Lindholm B, et al: High mobility group box protein-1 correlates with renal function in chronic kidney disease (CKD). Mol Med. 14:109–115. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Sato F, Maruyama S, Hayashi H, et al: High mobility group box chromosomal protein 1 in patients with renal diseases. Nephron Clin Pract. 108:c194–c201. 2008. View Article : Google Scholar : PubMed/NCBI

54 

Leelahavanichkul A, Huang Y, Hu X, et al: Chronic kidney disease worsens sepsis and sepsis-induced acute kidney injury by releasing high mobility group box protein-1. Kidney Int. 80:1198–1211. 2011. View Article : Google Scholar : PubMed/NCBI

55 

Nakamura T, Sato E, Fujiwara N, et al: Positive association of serum levels of advanced glycation end products and high mobility group box-1 with asymmetric dimethylarginine in nondiabetic chronic kidney disease patients. Metabolism. 58:1624–1628. 2009. View Article : Google Scholar

56 

D’Agati V and Schmidt AM: RAGE and the pathogenesis of chronic kidney disease. Nat Rev Nephrol. 6:352–360. 2010.

57 

Lynch J, Nolan S, Slattery C, Feighery R, Ryan MP and McMorrow T: High-mobility group box protein 1: a novel mediator of inflammatory-induced renal epithelial-mesenchymal transition. Am J Nephrol. 32:590–602. 2010. View Article : Google Scholar : PubMed/NCBI

58 

Sandim V, Pereira D, Ornellas A and Alves G: Renal cell carcinoma and proteomics. Urol Int. 84:373–377. 2010. View Article : Google Scholar : PubMed/NCBI

59 

Lin L, Zhong K, Sun Z, Wu G and Ding G: Receptor for advanced glycation end products (RAGE) partially mediates HMGB1-ERKs activation in clear cell renal cell carcinoma. J Cancer Res Clin Oncol. 13:11–22. 2012. View Article : Google Scholar : PubMed/NCBI

60 

Ganji MR and Harririan A: Chronic allograft dysfunction: major contributing factors. Iran J Kidney Dis. 6:88–93. 2012.PubMed/NCBI

61 

Wang S, Schmaderer C, Kiss E, et al: Recipient Toll-like receptors contribute to chronic graft dysfunction by both MyD88- and TRIF-dependent signaling. Dis Model Mech. 3:92–103. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April 2013
Volume 31 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhu P, Xie L, Ding H, Gong Q, Yang J and Yang L: High mobility group box 1 and kidney diseases (Review). Int J Mol Med 31: 763-768, 2013
APA
Zhu, P., Xie, L., Ding, H., Gong, Q., Yang, J., & Yang, L. (2013). High mobility group box 1 and kidney diseases (Review). International Journal of Molecular Medicine, 31, 763-768. https://doi.org/10.3892/ijmm.2013.1286
MLA
Zhu, P., Xie, L., Ding, H., Gong, Q., Yang, J., Yang, L."High mobility group box 1 and kidney diseases (Review)". International Journal of Molecular Medicine 31.4 (2013): 763-768.
Chicago
Zhu, P., Xie, L., Ding, H., Gong, Q., Yang, J., Yang, L."High mobility group box 1 and kidney diseases (Review)". International Journal of Molecular Medicine 31, no. 4 (2013): 763-768. https://doi.org/10.3892/ijmm.2013.1286