GET THE APP

From Humans to Experimental Models: The Cytoprotective Role of Cl
Medical & Surgical Urology

Medical & Surgical Urology
Open Access

ISSN: 2168-9857

+44-20-4587-4809

Review Article - (2014) Volume 3, Issue 2

From Humans to Experimental Models: The Cytoprotective Role of Clusterin in the Kidney

Qiunong Guan, Hatem A Alnasser, Christopher YC Nguan and Caigan Du*
Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
*Corresponding Author: Caigan Du, PhD, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada, Tel: 604-875-4111 Exn. 63793, Fax: 604- 875-5654 Email:

Abstract

Clusterin (CLU) is a chaperone-like protein and has been discovered more than thirty years ago; however, its biological significance is still not fully understood. This review aims to summarize the principal observations of CLU roles related to the kidney. In humans, three or more mRNA isoforms of CLU could be expressed due to different translation start sites, but only two forms of CLU protein, secreted (sCLU, isoform 2) and nuclear (nCLU, isoform 1), have been well characterized, whereas there is only sCLU form in mice. In the biopsies of renal tissue from patients, up regulated CLU expression has been found in rejecting kidney transplants or diseased kidneys, and a lower level of serum CLU is correlated with many types of kidney disease in patients. In mice, a deficiency in CLU expression specifically leads to the phenotype of age-dependent chronic glomerular injury - moderate to severe accumulation of the mesangial matrix, becomes more susceptible to ischemia-reperfusion injury (IRI), negatively impacts renal repair after IRI and worsens renal fibrosis after ureteral obstruction. All these observations may imply the biological significance of CLU for the maintenance of the tissue homeostasis in adult kidneys. However, how CLU protects the kidney from injury or by which extracellular and intracellular pathways mediate the cyto-protection of CLU in the kidney has not been well investigated. Understanding of the cyto-protective activities of CLU in the kidney could lead to the development of novel therapeutic strategies for the prevention and/or treatment of kidney injury or diseases.

<

Keywords: Clusterin, Extracellular chaperone, Kidney disease, Tissue homeostasis

CLU Gene, Isoforms and Cellular Localization

Clusterin (CLU) protein was first discovered more than thirty years ago [1], and a large volume of research has been dedicated to it since – there are more than two thousand publications in Pubmed/NCBI databases when using ‘clusterin’ as a keyword search criteria today. Human CLU gene (NCBI Gene ID: 1191) is located at chromosome 8p21-p12, and consists of 10 exons, in which the first two exons are alternative (designated 1 and 1’) [2]. Thus, CLU gene can be transcribed into at least three mRNA variants (NCBI Reference No.: NM_001831.3; NR_038335.1; NR_045494.1) or perhaps even more [3]. The mRNA isoform 1 is a major form of CLU mRNA, whereas other forms including mRNA isoform 2 collectively count for less than 1% of total CLU mRNA [3]. Two isoforms of CLU proteins have been well characterized; nuclear isoform of CLU (nCLU, isoform 1) containing the nuclear localization signal that is translated due to the splicing at exon 1 and 3 together placing a downstream AUG at exon 3 as the first available translation and lacking of exon 2 [3,4], while pre-secreted isoform of CLU (sCLU) containing the endoplasmic reticulum (ER)-targeting signalencoding in exon 2 [3]. The nCLU is translocated into the nucleus after translation andprobably without glycosylation [3], whereas the pre-secreted sCLU is targeted to ER and Golgi bodies glycosylation and cleavage between Arg-205 and Ser-206 to produce mature sCLU, a secreted disulfide-linked heterodimer of α- and β-chains [5-7]. Under certain stress conditions, sCLU however can be retrotranslocalized into the cytosol instead of secretion [8]. However, the cellular localization of all these isoforms and their expression are largely unknown. Murine CLU gene (NCBI Gene ID: 12759; MGI ID: 88423) is found at chromosome 14, and contains nine exons that are only transcribed to a single mRNA (NCBI Reference No.: NM_013492.2, 1808 bp) [9]. nCLU isoform has not been found in mice as of yet. The homolog of mouse CLU to human sCLU is 75% at the amino acid level, and both have the same ER-targeting signal peptide and the cleavage site [9] (Figure 1). By immunohistological staining using the same anti-CLU α-chain antibody, CLU protein was localized inhuman kidney sections in the same pattern as that of mice (Figure 2). Thus, CLU in mice can serve as a counterpart for sCLU (both extracellular and intracellular) in humans, particularly in the study of the kidney disorders.

medical-surgical-urology-sequence-alignment

Figure 1: Sequence alignment between mouse CLU (top sequence) and human sCLU (bottom sequence). The same amino acid residues were underlined with “”. ER-targeting signal peptide (mouse: 1-21; human: 1-22), and the cleavage site (Mouse: R226 – S227; human: R227-S228).

medical-surgical-urology-transplant-rejection

Figure 2: Induction of CLU protein in both tubules and glomeruli following transplant rejection or IRI. Kidney tissues were fixed in 10% buffered formaldehyde, embedded in paraffin, and then sectioned for immunohistochemical staining with goat polyclonal anti-CLU-α (C-18). (A) Biopsy sample of rejecting kidney transplant (Banff 97 grade 1 chronic rejection) from a 15-year old female patient (provided by Dr. Alex Magil, St. Paul’s Hospital, Vancouver, BC, Canada). (B) Mouse kidneys with renal ischemia-reperfusion injury. Brown color (also pointed by red arrows): positively stained cells. The data are presented as a typical image from each examination.

In the human body, sCLU is a major glycoprotein in all the physiological fluids such as plasma, milk, urine, cerebrospinal fluid, and semen [6]. It is constitutively produced and secreted by almost all cell types that form the cellular interfaces of fluid compartments [6], and similarly by the liver [10]. The serum levels of sCLU in humans are present in a range of 35-353 μg/mL [11-14]. In tissue, upregulation of CLU expression (probably including nCLU) is associated with many pathophysiological processes, such as neuropathologies [15,16], heart disease [17], cancer [18-20], kidney transplant rejection, and kidney disease including glomerulonephritis [21,22]. In rodents, renal CLU is upregulated following a variety of insults, such as unilateral ureteral obstruction (UUO) and ischemia-reperfusion injury (IRI) [23,24], acute glycerol-induced renal failure, chronic vitamin E and selenium deficiency [25], lupus-like nephritis [26], and in resident glomerular cells exposed to complement-mediated injury [27]. The CLU expression in glomerular mesangial and epithelial cells, as well as renal proximal tubular epithelial cell (TEC) is increased in response to the stimulation of thrombin [28] and hypoxia in our unpublished observations. Further studies indicate that CLU is an apically secreted glycoprotein in renal TECs [29], and is detected in both viable and apoptotic cells following renal injury [23,30]. The molecular mechanism(s) for either constitutive or inducible expression of CLU have not been well investigated. It has been reported that CLU gene proximal promoter contains a ‘clusterin element’ (CLE) that is specifically bound by heat-shock factor (HSF) 1 after heat shock, or by HSF1-HSF2 up on proteasome inhibition [31,32], resulting in the induction of CLU gene transcription.

Biochemistry and Functions of sCLU

The primary structure of sCLU in both α- and β-chain subunits contains several large molten globule domains, amphipathic regions and coiled-coil α-helices [9,33-35], which are typical of molecular chaperone - conformational adaptability to allow CLU protein to bind its substrate proteins or lipids with high affinity and low specificity [35-37]. Furthermore, one study has revealed that the glycosylation of sCLU may not be required for its overall secondary structure content and binding activity to its substrate [38].

The biological activities of extracellular sCLU have been studied extensively; it was initially found to induce cell aggregation [39-41], and in the plasma it inhibited the cytolysis of complement membrane attack complex (MAC) by binding to the complement components [42-44], and was associated with both high-density lipoprotein (HDL) and low-density lipoprotein (LDL) complexes [33,45,46]. These studies suggest that sCLU in the blood may serve not only as an inhibitor of the lytic terminal complement cascade, but also as a regulator of lipid transport and local lipid redistribution. Furthermore, addition of sCLU prevents cell apoptosis in cultured cells treated with TNF-α, H2O2 or gentamicin probably by activation of magalinphosphatidylinositol 3-kinase/Akt pathway [47-49], and mediates clearance of cellular debris into non-professional phagocytes [50]. Based on all of these observations, it has been proposed that sCLU functions as an extracellular chaperone, a previously unknown qualitycontrol system for protein folding that mediates the recognition and disposal of extracellular misfolded proteins via receptor (i.e. megalin)- mediated endocytosis and lysosomal degradation [51]. This hypothesis is supported by a recent study showing that sCLU in the blood binds to a panel of proteins, including ceruloplasmin, fibrinogen, and albumin, in response to physiologically relevant stress [52].

Inside human cells, in addition to nCLU isoform that is mainly localizing in the nucleus and triggers cell death [53], sCLU could redirect to the cytosol under cellular stress [8,54]. In mice, CLU in normal tissues (heart and kidney) is present as a single protein band at approximately 40 kDa in Western blot analysis in our studies [24,55,56], while in cultured cells from these tissues two protein bands at approximately 60 kDa and 40 kDa are detected [24,55,56], suggesting that mouse CLU probably is also retrotranslocated to the cytosol following exposure to sub lethal stress in culture conditions. A variety of biological activities of intracellular sCLU or cytoplasmic CLU (cCLU) have been reported; it inhibits apoptosis by the interaction with BAX or GRP78 [54,57,58] or promotes cell survival by the activation of Akt and NF-κB pathway [48,59]. It is of much more interest to see that sCLU (~70 kDa) acts as an intracellular chaperone to interact with both ATP7A and ATP7B (Cu-ATPases) and facilitates degradation of misfolded/mislocalized mutant ATP7B [60]. Whether or not intracellular sCLU plays a role in ER quality control machinery that facilitates the degradation of mis/ un-folded proteins in ER remains further investigation.

Requirement of CLU for organogenesis

During rodent embryogenesis, CLU expression is detected in a variety of the tissues in many developing organs, such as the epithelial cells of comma and S-shaped bodies of the primordial kidney [61], developing islet of Langerhans of the primordial pancreas [62], myocardial cells adjacent to developing endocardial cushions of both atrioventricular canal and truncusarteriosus, stromal connective tissue throughout leaflet formation of the developing hearts [63], hypothalamic region, neocortex and hippocampus of the developing brain [64,65]. Interestingly however, complete knockout (KO) of CLU expression in CLU KO mice has not been found to cause any phenotypic change in postnatal development as compared to WT mice [66], suggesting that CLU may not be absolutely required for the differentiation and morphogenesis of an organ (i.e. kidney). Although brain weight, neurons, astrocytes and oligodendrocytes are not significantly different between WT and CLU KO mice during postnatal development, it is noted that there is a significant deficit in motor cells (~16%) in the facial nucleus in CLU KO compared with WT mice [64], suggesting that CLU may have a negative impact on neuronal development in certain motor nuclei. Indeed, in cultured progenitor or undifferentiated cells, CLU enhances neuronal differentiation from neural precursor cells [67], and ectopic over expression of CLU significantly up-regulates the expression of morphogenic factor Pdx-1 and Ngn-3 that is correlated with an increase in β-cell transformation from neogenic ductal cells [68], and increases CXCR4 expression and migration of cardiac progenitor cells by [69]. We have recently demonstrated that kidney repair or tissue regeneration is impaired after IRI in CLU KO mice [55], suggesting that CLU may play a key role in the differentiation and migration of renal stem/progenitor cells that have been found to contribute to renal repair after injury [70-72] which however remain elusive.

CLU as a Kidney ‘Bodyguard’

The cytoprotection of sCLU in human kidney disease

sCLU in the body fluids has been reported to bind to MAC component(s) [42-44], all types of immunoglubins, particularly aggregated IgG [73], and lipoprotein particles [33,45,46]. In renal biopsies from all forms of kidney disease, the terminal complement complex is identified, at least partly, in sCLU-SC5b-9 complex both in the specific immune glomerular deposition and in the “non-specific” deposition in areas of renal injury [11,21,74,75], and glomerular CLU is co-localized with LDL receptor (LDR-R) in patients with membranous glomerulonephritis (MGN) and is associated with a reduction of proteinuria after a follow-up of 1.5 years [74]. In patients’ sera, sCLU levels are markedly lower with active membranous nephropathy (MN), focal segmental glomerulosclerosis (FSGS), and in children with steroid-responsive nephritic syndrome (NS) compared to controls [76]. So far, the role of sCLU in the pathogenesis of human kidney disease has not been well investigated. Saunders et al. [77] have reported that perfusion with sCLU-depleted plasma from patients with Heymann nephritis induces glomerular injury and significantly greater proteinuria in an isolated rat kidney model, and sCLU prevents MGN serum-activated cellular stress in cultured podocytes [74]. All these observations may uncover an important role for serum sCLU in the protection of the kidney fromplasma-induced injury, which however remains further investigation.

An emerging hypothesis suggests that extracellular chaperones (ECs) including sCLU likely patrol biological fluids for misfolded proteins and facilitate their clearance via endocytic receptors to maintain protein proteostasis in fluids such as plasma [51,78,79]. If this hypothesis is correct, sCLU may protect the kidney from injury by at least two mechanisms: First, extracellular sCLU may facilitate the clearance of mutant, misfolded or unfolded proteins that are the result of immune activation and/or direct nephrotoxicity. Hence, a lack of sCLU or the imbalance of sCLU to “nephrotoxic” proteins will result in the aggregation/deposition of these proteins in the kidney and cause kidney damage. Indeed, a high level of aberrantly glycosylated IgA1 and its associated immunocomplexes induce glomerular injury, and are a pathogenic factor for the development of IgA nephropathy [80-83], and extracellular mis-/unfolded amyloidogenic “precursor proteins”, such as serum amyloid A, apolipo protein AII, and Ig light/ heavy, form amyloid fibril deposition in the kidney, causing kidney injury and failure (amyloidosis-associated kidney disease) [84]. Second, extracellular sCLU may be required for the efficiency of the kidney in metabolic clearance of proteins. It has been recognized previously that the kidney is responsible for 30% to 80% of the metabolic clearance of protein/peptide ‘waste’ in the plasma [85,86], and recently, Wilson et al. has demonstrated that proteins injected into rats are cleared more rapidly from circulation when complexed with sCLU [79], suggesting that in low sCLU states, the inefficiency of the metabolic clearance of protein waste will induce cellular stress in the kidney, resulting in the disturbance of tissue homeostasis. Further experimental studies are needed to confirm this novel observation.

CLU and kidney injury in animal models

The effort to understand the role of sCLU in the pathogenesis of kidney disease using CLU KO mice has been carried out by our lab and others. Total knockout CLU expression in mice does not change their phenotype [66]; organ development and reproduction in young CLU KO mice are not different from wild type (WT) mice. However, by 21 months of age, up to 75% of glomeruli in CLU KO mice exhibit moderate to severe mesangial lesions - the accumulation of the mesangial matrix and the presence of intra mesangialtubulo-fibrillary structures as compared to little or no glomerular injury in WT controls [87]. Furthermore, the immune complexes of IgG, IgM, IgA, and in some cases C1q, C3, and C9 in the glomeruli could be detectable as early as 4 weeks of age of CLU KO mice, and these immune complex lesions can be induced as early as 3 months of age by unilateral nephrectomy [87]. The phenotype of age-dependent glomerular injury in CLU KO mice clearly suggests the biological significance of CLU for tissue homeostasis of the kidney. Recently, we and others have demonstrated that following renal IRI or UUO, renal CLU expression is up-regulated [24,55,88], and the lack of CLU expression in the kidneys worsens IRI [24], impairs renal tissue repair after IRI [55] and accelerates renal fibrosis or increases the levels of plasminogen activator inhibitor (PAI)-1, type I collagen, and fibronectin in response to obstruction [88]. These results may suggest that upregulation of CLU during renal injury is a protective response that may prevent cell death during IRI, facilitate renal tubular cell proliferation for renal repair after IRI, and maintain renal tissue homeostasis against the development of renal fibrosis. These observations suggest there is still much to learn about the role of sCLU in development various kidney pathologies. Further studies by using CLU KO mice as a negative control to investigate the cytoprotection of CLU in the kidneys of WT mice are needed.

Conclusion

The study of CLU in acute kidney injury and chronic kidney disease is but one segment of a host of additional biomedical research fields such as cancer, cardiovascular disease and Alzheimer’s disease, which are actively studying the role of CLU since it was discovered more than 30 years ago. Accumulating evidence in the literature reveals the chaperone activity of sCLU in both extracellular and intracellular fluids to maintain the protein proteostasis, by which sCLU could protect the kidney from injury (Figure 3). We believe that further understanding of the role of sCLU in the development of kidney disease is required and may help to develop therapeutic strategies specific for the prevention or treatment of a variety of renal pathological states.

medical-surgical-urology-physiological-stress

Figure 3: A simple scheme of hypothesis for the cytoprotection of sCLU in the kidney. Under normal condition or when physiological stress is minimal (+ CLU), sCLU in the plasma acts as a non-specific extracellular chaperone for all kind of mutant, misfolded and unfolded proteins, and prevents autoimmune response to and renal deposition of these ‘nephrotoxic’ proteins. Serum sCLU may also facilitate the protein clearance and intracellular sCLU maintains tissue homeostasis in the kidney. When the physiological stress is increased or under certain pathological condition (- CLU), misfolded and/or unfolded proteins are up-regulated both in the plasma and inside the cell. As a result, there is not enough sCLU to neutralize the accumulation of these proteins – leading to the immune activation and the deposition of these proteins in glomerular capillaries, and renal cell death via ER stress.

Acknowledgements

This study was supported by a grant of the Kidney Foundation of Canada. HAA was a graduate student supported by the scholarship from King Saud University (Riyadh, Saudi Arabia).

References

  1. Hogg SD, Embery G (1979) The isolation and partial characterization of a sulphated glycoprotein from human whole saliva which aggregates strains of Streptococcus sanguis but not Streptococcus mutans. Arch Oral Biol 24: 791-797.
  2. Tobe T, Minoshima S, Yamase S, Choi NH, Tomita M, et al. (1991) Assignment of a human serum glycoprotein SP-40,40 gene (CLI) to chromosome 8. Cytogenet Cell Genet 57: 193-195.
  3. Prochnow H, Gollan R, Rohne P, Hassemer M, Koch-Brandt C, Baiersdorfer M (2013) Non-secreted clusterin isoforms are translated in rare amounts from distinct human mRNA variants and do not affect Bax-mediated apoptosis or the NF-kappaB signaling pathway. PLoS One 8:e75303.
  4. Leskov KS, Klokov DY, Li J, Kinsella TJ, Boothman DA (2003) Synthesis and functional analyses of nuclear clusterin, a cell death protein. J BiolChem 278: 11590-11600.
  5. Burkey BF, deSilva HV, Harmony JA (1991) Intracellular processing of apolipoprotein J precursor to the mature heterodimer. J Lipid Res 32: 1039-1048.
  6. Jones SE, Jomary C (2002) Clusterin. Int J Biochem Cell Biol 34: 427-431.
  7. Kapron JT, Hilliard GM, Lakins JN, Tenniswood MP, West KA, et al. (1997) Identification and characterization of glycosylation sites in human serum clusterin. Protein Sci 6: 2120-2133.
  8. Nizard P, Tetley S, Le Dréan Y, Watrin T, Le Goff P, et al. (2007) Stress-induced retrotranslocation of clusterin/ApoJ into the cytosol. Traffic 8: 554-565.
  9. Jordan-Starck TC, Lund SD, Witte DP, Aronow BJ, Ley CA, et al. (1994) Mouse apolipoprotein J: characterization of a gene implicated in atherosclerosis. J Lipid Res 35: 194-210.
  10. Polihronis M, Machet D, Saunders J, O'Bryan M, McRae J, et al. (1993) Immunohistological detection of C5b-9 complement complexes in normal and pathological human livers. Pathology 25: 20-23.
  11. Murphy BF, Kirszbaum L, Walker ID, d'Apice AJ (1988) SP-40,40, a newly identified normal human serum protein found in the SC5b-9 complex of complement and in the immune deposits in glomerulonephritis. J Clin Invest 81: 1858-1864.
  12. Choi NH, Tobe T, Hara K, Yoshida H, Tomita M (1990) Sandwich ELISA assay for quantitative measurement of SP-40,40 in seminal plasma and serum. J Immunol Methods 131: 159-163.
  13. Morrissey C, Lakins J, Moquin A, Hussain M, Tenniswood M (2001) An antigen capture assay for the measurement of serum clusterin concentrations. J BiochemBiophys Methods 48: 13-21.
  14. Ståhl AL, Kristoffersson A, Olin AI, Olsson ML, Roodhooft AM, et al. (2009) A novel mutation in the complement regulator clusterin in recurrent hemolytic uremic syndrome. MolImmunol 46: 2236-2243.
  15. Polihronis M, Paizis K, Carter G, Sedal L, Murphy B (1993) Elevation of human cerebrospinal fluid clusterin concentration is associated with acute neuropathology. J NeurolSci 115: 230-233.
  16. Choi-Miura NH, Oda T (1996) Relationship between multifunctional protein "clusterin" and Alzheimer disease. Neurobiol Aging 17: 717-722.
  17. Trougakos IP, Poulakou M, Stathatos M, Chalikia A, Melidonis A, et al. (2002) Serum levels of the senescence biomarker clusterin/apolipoprotein J increase significantly in diabetes type II and during development of coronary heart disease or at myocardial infarction. ExpGerontol 37: 1175-1187.
  18. Trougakos IP, Gonos ES (2002) Clusterin/apolipoprotein J in human aging and cancer. Int J Biochem Cell Biol 34: 1430-1448.
  19. Zoubeidi A, Chi K, Gleave M (2010) Targeting the cytoprotective chaperone, clusterin, for treatment of advanced cancer. Clin Cancer Res 16: 1088-1093.
  20. Zoubeidi A, Gleave M (2012) Small heat shock proteins in cancer therapy and prognosis. Int J Biochem Cell Biol 44: 1646-1656.
  21. Murphy BF, Davies DJ, Morrow W, d'Apice AJ (1989) Localization of terminal complement components S-protein and SP-40,40 in renal biopsies. Pathology 21: 275-278.
  22. Dvergsten J, Manivel JC, Correa-Rotter R, Rosenberg ME (1994) Expression of clusterin in human renal diseases. Kidney Int 45: 828-835.
  23. Witzgall R, Brown D, Schwarz C, Bonventre JV (1994) Localization of proliferating cell nuclear antigen, vimentin, c-Fos, and clusterin in the postischemic kidney. Evidence for a heterogenous genetic response among nephron segments, and a large pool of mitotically active and dedifferentiated cells. J Clin Invest 93:2175-2188.
  24. Zhou W, Guan Q, Kwan CC, Chen H, Gleave ME, et al. (2010) Loss of clusterin expression worsens renal ischemia-reperfusion injury. Am J Physiol Renal Physiol 298: F568-578.
  25. Nath KA, Dvergsten J, Correa-Rotter R, Hostetter TH, Manivel JC, et al. (1994) Induction of clusterin in acute and chronic oxidative renal disease in the rat and its dissociation from cell injury. Lab Invest 71: 209-218.
  26. Moll S, Menoud PA, French L, Sappino AP, Pastore Y, et al. (1998) Tubular up-regulation of clusterin mRNA in murine lupus-like nephritis. Am J Pathol 152: 953-962.
  27. Yamada K, Hori Y, Hanafusa N, Okuda T, Nagano N, et al. (2001) Clusterin is up-regulated in glomerular mesangial cells in complement-mediated injury. Kidney Int 59: 137-146.
  28. Laping NJ, Olson BA, Short B, Albrightson CR (1997) Thrombin increases clusterin mRNA in glomerular epithelial and mesangial cells. J Am SocNephrol 8: 906-914.
  29. Graichen R, Lösch A, Appel D, Koch-Brandt C (1996) Glycolipid-independent sorting of a secretory glycoprotein to the apical surface of polarized epithelial cells. J BiolChem 271: 15854-15857.
  30. Gobé GC, Buttyan R, Wyburn KR, Etheridge MR, Smith PJ (1995) Clusterin expression and apoptosis in tissue remodeling associated with renal regeneration. Kidney Int 47: 411-420.
  31. Michel D, Chatelain G, North S, Brun G (1997) Stress-induced transcription of the clusterin/apoJ gene. Biochem J 328 : 45-50.
  32. Loison F, Debure L, Nizard P, le Goff P, Michel D, et al. (2006) Up-regulation of the clusterin gene after proteotoxic stress: implication of HSF1-HSF2 heterocomplexes. Biochem J 395: 223-231.
  33. de Silva HV, Harmony JA, Stuart WD, Gil CM, Robbins J (1990) Apolipoprotein J: structure and tissue distribution. Biochemistry 29: 5380-5389.
  34. de Silva HV, Stuart WD, Park YB, Mao SJ, Gil CM, et al. (1990) Purification and characterization of apolipoprotein J. J BiolChem 265: 14292-14297.
  35. Bailey RW, Dunker AK, Brown CJ, Garner EC, Griswold MD (2001) Clusterin, a binding protein with a molten globule-like region. Biochemistry 40: 11828-11840.
  36. Humphreys DT, Carver JA, Easterbrook-Smith SB, Wilson MR (1999) Clusterin has chaperone-like activity similar to that of small heat shock proteins. J BiolChem 274: 6875-6881.
  37. Nuutinen T, Suuronen T, Kauppinen A, Salminen A (2009) Clusterin: a forgotten player in Alzheimer's disease. Brain Res Rev 61: 89-104.
  38. Stewart EM, Aquilina JA, Easterbrook-Smith SB, Murphy-Durland D, Jacobsen C, et al. (2007) Effects of glycosylation on the structure and function of the extracellular chaperone clusterin. Biochemistry 46: 1412-1422.
  39. Fritz IB, Burdzy K, Sétchell B, Blaschuk O (1983) Ram rete testis fluid contains a protein (clusterin) which influences cell-cell interactions in vitro. BiolReprod 28: 1173-1188.
  40. Cheng CY, Mathur PP, Grima J (1988) Structural analysis of clusterin and its subunits in ram rete testis fluid. Biochemistry 27: 4079-4088.
  41. Silkensen JR, Skubitz KM, Skubitz AP, Chmielewski DH, Manivel JC, et al. (1995) Clusterin promotes the aggregation and adhesion of renal porcine epithelial cells. J Clin Invest 96: 2646-2653.
  42. Murphy BF, Saunders JR, O'Bryan MK, Kirszbaum L, Walker ID, et al. (1989) SP-40,40 is an inhibitor of C5b-6-initiated haemolysis. IntImmunol 1: 551-554.
  43. Choi NH, Nakano Y, Tobe T, Mazda T, Tomita M (1990) Incorporation of SP-40,40 into the soluble membrane attack complex (SMAC, SC5b-9) of complement. IntImmunol 2: 413-417.
  44. Tschopp J, Chonn A, Hertig S, French LE (1993) Clusterin, the human apolipoprotein and complement inhibitor, binds to complement C7, C8 beta, and the b domain of C9. J Immunol 151: 2159-2165.
  45. Jenne DE, Lowin B, Peitsch MC, Böttcher A, Schmitz G, et al. (1991) Clusterin (complement lysis inhibitor) forms a high density lipoprotein complex with apolipoprotein A-I in human plasma. J BiolChem 266: 11030-11036.
  46. Karlsson H, Leanderson P, Tagesson C, Lindahl M (2005) Lipoproteomics I: mapping of proteins in low-density lipoprotein using two-dimensional gel electrophoresis and mass spectrometry. Proteomics 5:551-565.
  47. Girton RA, Sundin DP, Rosenberg ME (2002) Clusterin protects renal tubular epithelial cells from gentamicin-mediated cytotoxicity. Am J Physiol Renal Physiol 282: F703-709.
  48. Ammar H, Closset JL (2008) Clusterin activates survival through the phosphatidylinositol 3-kinase/Akt pathway. J BiolChem 283: 12851-12861.
  49. Kim JH, Kim JH, Jun HO, Yu YS, Min BH, et al. (2010) Protective effect of clusterin from oxidative stress-induced apoptosis in human retinal pigment epithelial cells. Invest Ophthalmol Vis Sci 51: 561-566.
  50. Bartl MM, Luckenbach T, Bergner O, Ullrich O, Koch-Brandt C (2001) Multiple receptors mediate apoJ-dependent clearance of cellular debris into nonprofessional phagocytes. Exp Cell Res 271: 130-141.
  51. Wyatt A, Yerbury J, Poon S, Dabbs R, Wilson M (2009) Chapter 6: The chaperone action of Clusterin and its putative role in quality control of extracellular protein folding. Adv Cancer Res 104: 89-114.
  52. Wyatt AR, Wilson MR (2010) Identification of human plasma proteins as major clients for the extracellular chaperone clusterin. J BiolChem 285: 3532-3539.
  53. Bettuzzi S, Rizzi F (2009) Chapter 5: Nuclear CLU (nCLU) and the fate of the cell. Adv Cancer Res 104: 59-88.
  54. Zhang H, Kim JK, Edwards CA, Xu Z, Taichman R, et al. (2005) Clusterin inhibits apoptosis by interacting with activated Bax. Nat Cell Biol 7: 909-915.
  55. Nguan CY, Guan Q, Gleave ME, Du C (2014) Promotion of cell proliferation by clusterin in the renal tissue repair phase after ischemia-reperfusion injury. Am J Physiol Renal Physiol 306: F724-733.
  56. Li S, Guan Q, Chen Z, Gleave ME, Nguan CY, et al. (2011) Reduction of cold ischemia-reperfusion injury by graft-expressing clusterin in heart transplantation. J Heart Lung Transplant 30: 819-826.
  57. Wang C, Jiang K, Gao D, Kang X, Sun C, Zhang Q, et al (2013) Clusterin protects hepatocellular carcinoma cells from endoplasmic reticulum stress induced apoptosis through GRP78. PLoS One 8:e55981.
  58. Li N, Zoubeidi A, Beraldi E, Gleave ME (2013) GRP78 regulates clusterin stability, retrotranslocation and mitochondrial localization under ER stress in prostate cancer. Oncogene 32: 1933-1942.
  59. Zoubeidi A, Ettinger S, Beraldi E, Hadaschik B, Zardan A, et al. (2010) Clusterin facilitates COMMD1 and I-kappaB degradation to enhance NF-kappaB activity in prostate cancer cells. Mol Cancer Res 8: 119-130.
  60. Materia S, Cater MA, Klomp LW, Mercer JF, La Fontaine S (2011) Clusterin (apolipoprotein J), a molecular chaperone that facilitates degradation of the copper-ATPases ATP7A and ATP7B. J BiolChem 286: 10073-10083.
  61. French LE, Chonn A, Ducrest D, Baumann B, Belin D, Wohlwend A, et al (1993) Murineclusterin: molecular cloning and mRNA localization of a gene associated with epithelial differentiation processes during embryogenesis. J Cell Biol 122:1119-1130.
  62. Min BH, Jeong SY, Kang SW, Crabo BG, Foster DN, et al. (1998) Transient expression of clusterin (sulfated glycoprotein-2) during development of rat pancreas. J Endocrinol 158: 43-52.
  63. Witte DP, Aronow BJ, Dry JK, Harmony JA (1994) Temporally and spatially restricted expression of apolipoprotein J in the developing heart defines discrete stages of valve morphogenesis. DevDyn 201: 290-296.
  64. Charnay Y, Imhof A, Vallet PG, Hakkoum D, Lathuiliere A, et al. (2008) Clusterin expression during fetal and postnatal CNS development in mouse. Neuroscience 155: 714-724.
  65. O'Bryan MK, Cheema SS, Bartlett PF, Murphy BF, Pearse MJ (1993) Clusterin levels increase during neuronal development. J Neurobiol 24: 421-432.
  66. McLaughlin L, Zhu G, Mistry M, Ley-Ebert C, Stuart WD, et al. (2000) Apolipoprotein J/clusterin limits the severity of murine autoimmune myocarditis. J Clin Invest 106: 1105-1113.
  67. Cordero-Llana O, Scott SA, Maslen SL, Anderson JM, Boyle J, et al. (2011) Clusterin secreted by astrocytes enhances neuronal differentiation from human neural precursor cells. Cell Death Differ 18: 907-913.
  68. Kim SY, Lee S, Min BH, Park IS (2007) Functional association of the morphogenic factors with the clusterin for the pancreatic beta-cell differentiation. Diabetes Res ClinPract 77 Suppl 1: S122-126.
  69. Li Y, Qu J, Shelat H, Gao S, Wassler M, et al. (2010) Clusterin induces CXCR4 expression and migration of cardiac progenitor cells. Exp Cell Res 316: 3435-3442.
  70. Romagnani P, Lasagni L, Remuzzi G (2013) Renal progenitors: an evolutionary conserved strategy for kidney regeneration. Nat Rev Nephrol 9: 137-146.
  71. Morigi M, Benigni A (2013) Mesenchymal stem cells and kidney repair. Nephrol Dial Transplant 28: 788-793.
  72. Smeets B, Boor P, Dijkman H, Sharma SV, Jirak P, et al. (2013) Proximal tubular cells contain a phenotypically distinct, scattered cell population involved in tubular regeneration. J Pathol 229: 645-659.
  73. Wilson MR, Easterbrook-Smith SB (1992) Clusterin binds by a multivalent mechanism to the Fc and Fab regions of IgG. BiochimBiophysActa 1159: 319-326.
  74. Rastaldi MP, Candiano G, Musante L, Bruschi M, Armelloni S, et al. (2006) Glomerular clusterin is associated with PKC-alpha/beta regulation and good outcome of membranous glomerulonephritis in humans. Kidney Int 70: 477-485.
  75. Sethi S, Gamez JD, Vrana JA, Theis JD, Bergen HR 3rd, et al. (2009) Glomeruli of Dense Deposit Disease contain components of the alternative and terminal complement pathway. Kidney Int 75: 952-960.
  76. Ghiggeri GM, Bruschi M, Candiano G, Rastaldi MP, Scolari F, et al. (2002) Depletion of clusterin in renal diseases causing nephrotic syndrome. Kidney Int 62: 2184-2194.
  77. Saunders JR, Aminian A, McRae JL, O'Farrell KA, Adam WR, et al. (1994) Clusterin depletion enhances immune glomerular injury in the isolated perfused kidney. Kidney Int 45: 817-827.
  78. Wyatt AR, Yerbury JJ, Ecroyd H, Wilson MR (2013) Extracellular chaperones and proteostasis. Annu Rev Biochem 82: 295-322.
  79. Wyatt AR, Yerbury JJ, Berghofer P, Greguric I, Katsifis A, et al. (2011) Clusterin facilitates in vivo clearance of extracellular misfolded proteins. Cell Mol Life Sci 68: 3919-3931.
  80. Coppo R, Amore A (2004) Aberrant glycosylation in IgA nephropathy (IgAN). Kidney Int 65: 1544-1547.
  81. Camilla R, Suzuki H, Daprà V, Loiacono E, Peruzzi L, et al. (2011) Oxidative stress and galactose-deficient IgA1 as markers of progression in IgA nephropathy. Clin J Am SocNephrol 6: 1903-1911.
  82. Zhao N, Hou P, Lv J, Moldoveanu Z, Li Y, et al. (2012) The level of galactose-deficient IgA1 in the sera of patients with IgA nephropathy is associated with disease progression. Kidney Int 82: 790-796.
  83. Tanaka M, Seki G, Someya T, Nagata M, Fujita T (2011) Aberrantly glycosylated IgA1 as a factor in the pathogenesis of IgA nephropathy. ClinDevImmunol 2011: 470803.
  84. Dember LM (2006) Amyloidosis-associated kidney disease. J Am SocNephrol 17: 3458-3471.
  85. Carone FA, Peterson DR, Oparil S, Pullman TN (1979) Renal tubular transport and catabolism of proteins and peptides. Kidney Int 16: 271-278.
  86. Maack T, Johnson V, Kau ST, Figueiredo J, Sigulem D (1979) Renal filtration, transport, and metabolism of low-molecular-weight proteins: a review. Kidney Int 16: 251-270.
  87. Rosenberg ME, Girton R, Finkel D, Chmielewski D, Barrie A 3rd, et al. (2002) Apolipoprotein J/clusterin prevents a progressive glomerulopathy of aging. Mol Cell Biol 22: 1893-1902.
  88. Jung GS, Kim MK, Jung YA, Kim HS, Park IS, et al. (2012) Clusterin attenuates the development of renal fibrosis. J Am SocNephrol 23: 73-85.
Citation: Guan Q, Alnasser HA, Nguan CYC, Du C (2014) From Humans to Experimental Models: The Cytoprotective Role of Cluster in the Kidney. Med Surg Urol 3:134.

Copyright: © 2014 Guan Q, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top