Skip to main content
Erschienen in: Diabetology & Metabolic Syndrome 1/2015

Open Access 01.12.2015 | Research

The role of progranulin in diabetes and kidney disease

verfasst von: Bruna Bellincanta Nicoletto, Luis Henrique Canani

Erschienen in: Diabetology & Metabolic Syndrome | Ausgabe 1/2015

Abstract

Progranulin (PGRN) is a cysteine rich secreted protein, expressed in epithelial cells, immune cells, neurons, and adipocytes. It was first identified for its growth factor-like properties, being involved in early embryogenesis and tissue remodeling, acting as an anti-inflammatory molecule. In the central nervous system, PGRN has neurotrophic and neuroprotective actions. There is also evidence of PGRN effects on cancer, contributing to tumor proliferation, invasion and cell survival. Recently, PGRN was recognized as an adipokine related to obesity and insulin resistance, revealing its metabolic function and pro-inflammatory properties. In obesity and type 2 diabetes mellitus, PGRN levels are increased. In renal disease, there is a relevant association, however, it is not known if it could contribute to kidney damage or if it is only a route of PGRN elimination. PGRN is an emerging molecule which demands studies in different fields. Possibly, it plays distinct functions in different tissues/cells and metabolic conditions. Here, we discuss potential mechanisms and recent data of PGRN pro-inflammatory actions, regarding obesity, insulin resistance, type 2 diabetes mellitus and kidney disease.
Abkürzungen
PGRN
progranulin
ERK
extracellular regulated kinase
PI3 K
phosphatidylinositol 3-kinase
FTD
frontotemporal dementia
TNFR
tumor necrosis factor receptor
TNFα
tumor necrosis factor-α
BMI
body mass index
MCP-1
monocyte chemoattractant protein-1
CRP
C-reactive protein
IL-6
interleukin 6
HOMA-IR
Homeostasis Model Assessment for insulin resistance
SOCS3
cytokine signaling-3
IRS-1
insulin receptor substrate-1
ER
endoplasmatic reticulum
mTOR
mammalian target of rapamycin
PPARg
peroxisome proliferator-activated receptor-gamma
T2DM
type 2 diabetes mellitus
CKD
chronic kidney disease
GFR
glomerular filtration rate
UAE
urinary albumin excretion
DKD
diabetic kidney disease
T1DM
type 1 diabetes mellitus
ERFD
early renal function decline
NOD2
nucleotide-binding oligomerization domain containing 2

Background

Progranulin (PGRN) is a 68–88 kDa cysteine rich secreted protein, also known as granulin-epithelin precursor, proepithelin or PC-cell derived growth factor [1, 2]. It is encoded by GRN (PGRN gene) and expressed in many cell types, including epithelial cells, immune cells, neurons, and adipocytes [3]. In kidneys, PGRN is expressed by renotubular epithelia of mouse embryo [4]. In healthy adult rodents, PGRN continues to be expressed in the kidney, strongly in the transitional epithelium of the ureter; but weakly in the proximal and distal convoluted tubules of the cortex and collecting ducts of the medulla [5]. In humans, the PGRN expression in kidneys remains unknown.
The first evidence of the protein was found during guinea pig spermatogenesis, when an acrosomal glycoprotein, named acrogranin, was detected [6]; and later identified as the guinea pig equivalent of PGRN [7]. PGRN has growth factor–like properties, being involved in early embryogenesis [4], wound repair and tissue remodeling [8]. It regulates cell division, survival, and migration, mainly via extracellular regulated kinase (ERK) and phosphatidylinositol 3-kinase (PI3K) pathways [8]. The growth factor–like properties of PGRN could be involved in physiology of tissue repair or in diseases such as cancer [1]. By the same pathways (ERK and PI3K), PGRN contributes to tumor proliferation, invasion and cell survival [911]. This molecule has previously been linked to many cancer types, as breast [12], ovarian [13], cervical [14], gastrointestinal [15] and kidney cancers [16].
Progranulin is secreted in an intact form and can be cleaved into granulins by proteases [2, 3]. Granulins are small proteins of approximately 6 kDa characterized by a conserved motif of 12 cysteines and play a role in the extracellular regulation of cell function and growth [8]. It has been suggested that the full length form of the protein (PGRN) has anti-inflammatory action, while released granulins have the opposite effect, increasing the production of proinflammatory cytokines, as interleukin 8 [1]. The intact PGRN is reported bind to tumor necrosis factor receptor (TNFR), inhibiting the binding of tumor necrosis factor-α (TNFα) and its proinflammatory signaling [17, 18]. In mouse models of arthritis, PGRN prevents inflammation [17]. In humans, increased serum levels of PGRN are observed in rheumatoid arthritis, but its relation to the pathogenesis of the disease remains unclear [19]. Elevated PGRN levels are observed in the skin of psoriasis patients [20, 21], mice dermatitis model [20, 21] and wounds [1]. Some authors suggest that PGRN has the effect of attenuating inflammation in these conditions, acting as an anti-inflammatory molecule [20, 21].
In central nervous system, PGRN has neurotrophic and neuroprotective actions [22]. It is involved in neurite outgrowth and possibly plays a role in plasticity and remodeling in the adult brain [23]. In addition, PGRN protects neurons from premature death [8] and acts in the response to stress [23] and neuroinflammation [22]. PGRN deficiency is associated with neurodegeneration and frontotemporal dementia (FTD), mainly due to mutation in GRN [24]. However, PGRN expression is upregulated in microglia in neurodegenerative disease [25] as FTD, especially in brain areas with a substantial pathology [26]. It is unclear if it represents a result of microglia response to injury or an active contribution to the disease progression [22].
After acute ischemia–reperfusion injury, lower PGRN levels are observed in mice brain [27] and kidney [28]; and treatment with recombinant PGRN could attenuate inflammation in this condition [27, 28]. PGRN also seems to protect against acute focal cerebral ischemia in rats by attenuation of blood–brain barrier disruption, neuroinflammation suppression, and neuroprotection [29].
Despite the reported anti-inflammatory properties of PGRN in some conditions, it seems to be a more complex molecule, revealing an opposite metabolic function. In the periphery, the intact form of PGRN has been associated with proinflammatory effects, since PGRN was recently recognized as an adipokine related to obesity and insulin resistance [3, 30]. Nothing further is known about the relationship of PGRN with its proteolytic granulins in obesity and insulin resistance.

Review

Adipose tissue, obesity and PGRN

Since the discovery of leptin in 1994, adipose tissue has been recognized as an endocrine organ, with its secreted adipokines playing many functions in the body [31]. Leptin acts on energy metabolism, regulating appetite and food intake, as an anorexigenic hormone [32]. In obesity, its levels are increased; however, leptin resistance is observed, impairing leptin functions [33]. A similar biological process is suggested for PGRN [34]. There is evidence that the administration of PGRN in the mice hypothalamus significantly suppresses fasting-induced feeding and body weight gain in a dose-dependent manner, possibly through hypothalamic neuropeptide Y and the melanocortin system [34]. However, in obesity, a resistance to the anorexigenic effects of PGRN may contribute to increased food intake [34].
In obesity, PGRN levels are increased [30, 35, 36]. Both experimental studies [35] and those performed in humans [30, 36] have reported the relationship between PGRN and adiposity. In ob/ob mice, a well-characterized obese and insulin resistance model, there are elevated PGRN serum levels and upregulation of Grn in white adipose tissue [35]. In humans, previous studies report a positive correlation between body mass index (BMI) and PGRN serum levels [30, 36, 37].
Progranulin is positively correlated to body fat percentage and waist circumference [3638]. Fat distribution is related to PGRN levels, revealing higher PGRN serum concentration in subjects with visceral obesity [36]. It is known that visceral fat and its secreted adipokines and immune cell-derived cytokines are involved in chronic inflammation [31]. PGRN seems to play a role on this process, due its chemotactic activity, recruiting monocytes into adipose tissue as well as monocyte chemoattractant protein-1 (MCP-1) [36]. Furthermore, there is evidence that Grn deficient mice had significantly less infiltration of macrophages in adipose tissue [35]. Taken together, these findings suggest a proinflammatory effect of PGRN. This is supported by human studies that found a positive correlation between PGRN and C-reactive protein (CRP) [36, 39] and interleukin 6 (IL-6) [30, 39].
The effects of PGRN in obesity and inflammation have also revealed its influence on insulin resistance. A positive correlation between PGRN levels and HOMA-IR index (Homeostasis Model Assessment for insulin resistance) has been reported [30, 37]. In morbid obesity, patients with insulin resistance have elevated PGRN serum concentration [40].
Experimental studies reported that PGRN promotes IL-6 expression in adipose cells, and its elevation enhances cytokine signaling-3 (SOCS3) expression via activation of JAK-STAT signaling. This mechanism can inhibit tyrosine phosphorylation of insulin receptor substrate-1 (IRS-1), leading to insulin resistance [35]. Additional evidence reports that Grn deficient mice fed with high fat diet presented improved insulin sensitivity. Moreover, body weight, fat mass and size of adipocytes were lower in Grn deficient mice compared to the wild-type mice receiving a standard diet [35]. These findings suggest a relevant association of PGRN with obesity and insulin resistance [35], as summarized in Fig. 1.
Recently, experimental studies reported that PGRN increases autophagic activity and triggers endoplasmatic reticulum (ER) stress in cultured human adipocytes, impacting on insulin signaling [37]. First, in multiple insulin-resistant cellular models there were increased levels of PGRN and autophagic imbalance. In PGRN deficient adipocytes, decreased markers of autophagy were observed. Moreover, adipocytes treated with PGRN and then stimulated with insulin revealed diminished IRS-1 and Akt phosphorylation and increased autophagic disorder. Mechanisms suggested involve ERK and mammalian target of rapamycin (mTOR) pathways [37]. ERK activation impairs IRS-1 activity and is associated with autophagy and ER stress-induced insulin resistance [41]. Likewise, inhibition of mTOR reduces insulin action and promotes autophagic disorders in adipocytes [42]. Possibly, PGRN effects are mediated by ERK activation and impaired mTOR phosphorylation [37] (Fig. 1). Moreover, there is evidence that PGRN could exert a causative role in hepatic insulin resistance, as observed in mice treated with PGRN for 21 days. Animals presented impaired glucose and insulin tolerance, and hepatic autophagy imbalance [43].
Regulation of PGRN on autophagy disorders and insulin resistance seems to be partially mediated through TNFR-1 via NF-kB signaling [37, 43]. Previous studies revealed that PGRN binds to TNFR, impairing the TNFα/TNFR interaction and suppressing chronic inflammation in mouse models of arthritis [17]. Although one study failed to demonstrate the binding of PGRN to TNFR [44], a recent publication reinforces this interaction [45]. Further evidence is required to elucidate the effects of PGRN binding to TNFR in different tissues, but it is possible that PGRN has dual roles in inflammation, exhibiting pro- and anti-inflammatory proprieties.
Some authors suggest that PGRN could be involved in the growth of adipose tissue [3]. It is known that the expansion of fat mass during obesity is followed by angiogenesis [46]; and PGRN has previously been linked to vessel formation [47]. Therefore, this adipokine could also contribute to increase adiposity (Fig. 1). However, this hypotheses needs to be confirmed by further studies.
Regarding adipogenesis, the role of PGRN is not fully understood. Its anti-adipogenic effects were reported in a previous study using porcine preadipocytes [48]. PGRN promoted ERK activation that phosphorylate peroxisome proliferator-activated receptor-gamma (PPARg) at the serine 112 site, impairing its function on adipocyte differentiation [48]. The effect of PGRN on ER stress also suppressed adipogenesis in cultured human adipose cells [37]. Moreover, Matsubara et al. [35] found that PGRN expression decreased with differentiation of 3T3-L1 adipose cells assessed by pre-adipocyte factor-1, PPARg, and fatty acid binding protein. On the other hand, a positive significant correlation between circulating PPARg and PGRN was observed in obese subjects [49].

Type 2 diabetes mellitus and PGRN

From 1988 to 2010, the total number of persons with diabetes increased by almost 75 % [50]. In 2012, there were an estimated 371 million adults living with diabetes, with 4.8 million deaths attributable to this disease [51].
Type 2 diabetes mellitus (T2DM) is characterized by a resistance to insulin action and inadequate compensatory insulin secretory response, leading to hyperglycemia [52]. Obesity and visceral fat are probably the main risk factors for T2DM [53] and are involved in its pathophysiology as well as inflammation and insulin resistance [31]. Genetic and T2DM family history are widely studied and several genes have been associated with T2DM development [54, 55].
The PGRN gene is located in the chromosome region 17q21.32. The long arm of chromosome 17 was previously linked to visceral adipose tissue, waist circumference and BMI in Hispanic families in the Insulin Resistance Atherosclerosis Family Study [56]. Moreover, linkage signals of fasting glucose was found with 17q in young European sib-pairs, suggesting an association with T2DM [57]. Other important genes are located in this chromosome region, as SOCS3 [58] and genes involved in food intake [59].
Recently, new markers have been studied in the pathogenesis of diabetes, involving many adipokines [60, 61], such as PGRN [30, 36, 38]. There is evidence that PGRN levels are increased in T2DM when compared to non-diabetic subjects [30, 36, 38, 62]. PGRN is closely related to glucose metabolism. There is a positive correlation between PGRN and A1C, fasting plasma glucose and 2 h post-challenge plasma glucose [30, 36, 37]. Elevated PGRN concentrations are also observed in impaired glucose tolerance subjects, revealing its role in prediabetic states [38]. Moreover, a recent study evaluating T2DM patients reports that obese subjects present higher levels of PGRN [30].
The association of PGRN with T2DM is mainly explained by its role in adipose tissue and insulin resistance. PGRN promotes IL-6 expression, impairing insulin signaling [35]. Moreover, it is a chemoattractant protein that recruits monocytes into adipose tissue, promoting inflammatory response with increased cytokines levels [36] (Fig. 1).
Decrease in circulating PGRN levels can be obtained with long term diet intervention [63] and exercise training [36]. A recent study evaluated the change in PGRN levels after 24 months of dietary intervention, and showed that the decrease was sustained throughout this period, irrespective of weight stabilization or partial weight regain [63]. Another study identified a significantly decrease of ~20 % in PGRN serum concentration after a 4-week training program, only in T2DM patients [36].
Other metabolic disorders associated with T2DM have also been linked to PGRN. A positive correlation observed between total cholesterol [36], triglycerides [30, 37] and PGRN suggests a role in dyslipidemia. Patients with metabolic syndrome present higher serum PGRN concentration [37, 64] and the number of metabolic syndrome components have a significant positive correlation with PGRN levels [39]. Elevation of PGRN expression in omental adipose tissue is also observed in patients with metabolic syndrome, indicating a potential contribution of adiposity to increased PGRN serum levels [37]. Moreover, patients with metabolic syndrome also present increased autophagic activity and ER stress in adipose tissue [37]. Finally, the effects of PGRN in obesity, insulin resistance and inflammation contribute to its association with atherosclerosis [39].
T2DM is associated with poor outcomes, characterized by macro and microvascular complications. Intensity and duration of hyperglycemia exposure leads to vascular and nervous damage, resulting in organ dysfunction, such as kidney, eyes, nerves, heart and blood vessels [52, 65].

Kidney disease and PGRN

Chronic kidney disease (CKD) is defined as a reduced glomerular filtration rate (GFR), increased urinary albumin excretion (UAE), or both. The incidence and prevalence of CKD differ substantially across countries; however, the estimated worldwide prevalence is around 8–16 % [66].
Although hypertension, glomerulonephritis and other comorbidities can lead to development of renal dysfunction, diabetes is the main cause of CKD [66]. In Brazil, it is the primary kidney disease in most patients starting dialysis [67]. The prevalence of diabetic kidney disease (DKD) increases over the years after the T2DM diagnosis, affecting about 25 % of patients with 10 years of the pathology [68]. Hyperglycemia associated with hypertension can lead to glomerulus injury [69, 70]. Tissue inflammation promotes kidney fibrosis, leading to protein clearance, such as albuminuria [71]. Although increased UAE are common in DKD, some patients with T2DM present reduced GFR, even in the absence of albuminuria [69, 70, 72].
Chronic kidney disease complications include increased all-cause and cardiovascular mortality, kidney-disease progression, acute kidney injury, anaemia, mineral and bone disorders, fractures and cognitive decline [66]. When associated with diabetes, an increased risk of mortality is observed [67, 73].
Recently, PGRN was described as an adipokine dependent of renal function [74]. Five hundred thirty-two patients with stages 1–5 of CKD (according to the National Kidney Foundation classification) had their PGRN serum levels evaluated. Even after adjustment for age, sex and BMI, PGRN remained significantly different between the five subgroups of CKD, being higher in stage 5. Moreover, estimated GFR was identified as an independent predictor of PGRN circulating levels. These findings suggest that renal filtration is an important route of PGRN elimination [74]. However, the authors did not find a significant correlation between serum and urinary PGRN in a subgroup of 145 patients, which limits their conclusions. There is also an hypothesis that PGRN might contribute to the proinflammatory state frequently observed in renal disease [74].
Progranulin also seems to be involved in DKD. In a recent study evaluating eighty-four T2DM patients, increased PGRN serum levels were described in macroalbuminuric subjects [75]. The study also evaluated the presence of proliferative diabetic retinopathy and observed higher levels of PGRN in this group of patients, suggesting PGRN as a marker for diabetic microangiopathy and its severity [75].
The association of urinary PGRN levels and renal damage was investigated in seventy-four patients with type 1 diabetes mellitus (T1DM) [76]. Subjects were evaluated at baseline, when urine was collected, and after 6 years, when albuminuria and early renal function decline (ERFD, defined as a decline in cystatin C-based estimated GFR of ≥3.3 % per year) were assessed. Patients with both ERFD and albuminuria presented higher urinary PGRN levels at baseline than patients who maintained normal renal function and normoalbuminuria, when adjusted by age, diabetes duration, baseline albumin excretion rate, HbA1C, cystatin C and uric acid. Moreover, PGRN was significantly predictive of ERFD and albuminuria in patients with type 1 diabetes in multivariable logistic regression [76]. The study also investigated urinary levels of Tamms–Horsfall glycoprotein, clusterin and human α-1 acid glycoprotein; and concludes that a panel of these three proteins plus PGRN could be used to predict early signs of DKD [76]. Table 1 summarizes present data regarding PGRN and renal function.
Table 1
Studies characteristics regarding PGRN and renal function
Characteristic/reference
Xu et al. [75]
Richter et al. [74]
Schlatzer et al. [76]
Patients
84 patients with T2DM and 12 health persons
532 patients with stages 1–5 of CKD
74 patients with T1DM
PGRN material
Serum
Serum
Urine
Design
Cross-sectional study
Cross-sectional study
Longitudinal study
Baseline: urine collection, PGRN dosage
3 and 6-year visit: assessment of MA and ERFD
Results regarding PGRN
PGRN serum levels are increased in T2DM patients with macroalbuminuria
Positive correlation between serum PGRN and urinary albumin excretion rate
Negative correlation between PGRN and eGFR
PGRN serum levels are different between groups of CKD stages
↑ PGRN levels at stage 5 of CKD
CKD stage or eGFR are independently associated with PGRN serum levels
Lowest PGRN levels in patients who maintained normal renal function and normoalbuminuria (n = 35)
Nonsignificant increase in patients with either ERFD (n = 15) or MA (n = 16)
Significant increase in patients with both ERFD and MA (n = 8)
Urinary PGRN was significantly predictive of ERFD and MA in patients with T1DM
Conclusion
PGRN might be considered as a marker for diabetic microangiopathy and its severity
Renal function assessed as eGFR is a strong, independent predictor of serum PGRN
PGRN serum levels significantly increase with deteriorating renal function assessed as CKD stage
A panel of 4 proteins (PGRN, Tamms-Horsfall glycoprotein, clusterin and human α-1 acid glycoprotein) could be used to predict early signs of DKD
CKD chronic kidney disease, T1DM type 1 diabetes mellitus, PGRN progranulin, ERFD early renal function decline, eGFR estimated glomerular filtration rate, MA micro- or macroalbuminuria, DKD diabetic kidney disease
There is little evidence regarding the association of PGRN and DKD in T2DM patients. The proinflammatory effects of this adipokine could be involved in the pathway of renal damage, decreasing GFR and increasing albuminuria. When CKD is established, PGRN clearance is reduced and its effects could be potentiated. However, further studies are needed to elucidate this hypothesis.
In an acute condition, such as renal ischemia–reperfusion injury, an experimental study observed lower levels of PGRN in the mice kidney [28]. Moreover, Grn deficient mice presented a higher elevation of serum creatinine and blood urea nitrogen, more severe morphological injury and higher inflammatory response. Administration of recombinant PGRN in vitro could attenuate inflammation after renal ischemia–reperfusion injury at least in part associated with a nucleotide-binding oligomerization domain containing 2 (NOD2)-mediated immune response [28]. Therefore, PGRN plays a protective role and has an anti-inflammatory effect in the kidney after renal ischemia–reperfusion injury [28].

Conclusions

Progranulin is an emerging molecule which demands studies in different fields. Previous data have identified PGRN as a pro- and anti-inflammatory protein. Possibly, it plays distinct functions in different tissues/cells and metabolic conditions, as reported in Table 2. It was previously demonstrated that expression of PGRN in intact skin is low, but in injured skin, it raises significantly [1]. In addition, PGRN exerts anorexigenic effect in lean state, but a resistance is observed in obesity, leading to increased food intake [34]. Moreover, in acute condition of ischemia–reperfusion injury, PGRN plays an anti-inflammatory effect [2729], while in obesity (a chronic condition), it is associated with insulin resistance and inflammation [35].
Table 2
Metabolic conditions associated with pro- or anti-inflammatory effects of PGRN
Proinflammatory
Anti-inflammatory
Obesity
Insulin resistance
T2DM
Dyslipidemia
Metabolic syndrome
Wound repair
Psoriasis
Central nervous system
Arthritis
Acute ischemia–reperfusion injury
T2DM type 2 diabetes mellitus
It is not fully understood if PGRN is a cause or consequence of some conditions. PGRN could be involved in the pathogenesis of obesity and T2DM, and become a target for metabolic disorders prevention or treatment. In renal disease, it is not known if it could contribute to kidney damage or if it is only a route of PGRN elimination. In the last case, PGRN could be used as a marker of renal disease. Further studies are necessary to elucidate these questions and investigate the crosstalk between pro- and anti-inflammatory PGRN proprieties in different tissues and conditions, in order to clarify the action mechanisms of this potential molecule.

Authors’ contributions

BBN and LHC wrote the manuscript. Both authors read and approved the final manuscript.

Acknowledgements

This work was performed at Post Graduation Medical Sciences Program: Endocrinology, School of Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil. Financial sponsorship was received from Centro de Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) and Fundo de Incentivo a Pesquisa e Eventos do Hospital de Clínicas de Porto Alegre (FIPE/HCPA).

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat He ZH, Bateman A. Progranulin (granulin-epithelin precursor, PC-cell-derived growth factor, acrogranin) mediates tissue repair and tumorigenesis. J Mol Med (Berl). 2003;81:600–12.PubMedCrossRef He ZH, Bateman A. Progranulin (granulin-epithelin precursor, PC-cell-derived growth factor, acrogranin) mediates tissue repair and tumorigenesis. J Mol Med (Berl). 2003;81:600–12.PubMedCrossRef
2.
Zurück zum Zitat Zhou J, Gao G, Crabb JW, Serrero G. Purification of an autocrine growth-factor homologous with mouse epithelin precursor from a highly tumorigenic cell-line. J Biol Chem. 1993;268:10863–9.PubMed Zhou J, Gao G, Crabb JW, Serrero G. Purification of an autocrine growth-factor homologous with mouse epithelin precursor from a highly tumorigenic cell-line. J Biol Chem. 1993;268:10863–9.PubMed
3.
Zurück zum Zitat Nguyen AD, Nguyen TA, Marten LH, Mitic LL, Farese RV Jr. Progranulin: at the interface of neurodegenerative and metabolic diseases. Trends Endocrinol Metab. 2013;24:597–606.PubMedCrossRef Nguyen AD, Nguyen TA, Marten LH, Mitic LL, Farese RV Jr. Progranulin: at the interface of neurodegenerative and metabolic diseases. Trends Endocrinol Metab. 2013;24:597–606.PubMedCrossRef
4.
Zurück zum Zitat Daniel R, Daniels E, He ZH, Bateman A. Progranulin (acrogranin/PC cell-derived growth factor/granulin-epithelin precursor) is expressed in the placenta, epidermis, microvasculature, and brain during murine development. Dev Dyn. 2003;227:593–9.PubMedCrossRef Daniel R, Daniels E, He ZH, Bateman A. Progranulin (acrogranin/PC cell-derived growth factor/granulin-epithelin precursor) is expressed in the placenta, epidermis, microvasculature, and brain during murine development. Dev Dyn. 2003;227:593–9.PubMedCrossRef
5.
Zurück zum Zitat Daniel R, He ZH, Carmichael KP, Halper J, Bateman A. Cellular localization of gene expression for progranulin. J Histochem Cytochem. 2000;48:999–1009.PubMedCrossRef Daniel R, He ZH, Carmichael KP, Halper J, Bateman A. Cellular localization of gene expression for progranulin. J Histochem Cytochem. 2000;48:999–1009.PubMedCrossRef
6.
Zurück zum Zitat Anakwe OO, Gerton GL. Acrosome biogenesis begins during meiosis—evidence from the synthesis and distribution of an acrosomal glycoprotein, acrogranin, during guinea-pig spermatogenesis. Biol Reprod. 1990;42:317–28.PubMedCrossRef Anakwe OO, Gerton GL. Acrosome biogenesis begins during meiosis—evidence from the synthesis and distribution of an acrosomal glycoprotein, acrogranin, during guinea-pig spermatogenesis. Biol Reprod. 1990;42:317–28.PubMedCrossRef
7.
Zurück zum Zitat Baba T, Hoff HB, Nemoto H, Lee H, Orth J, Arai Y, et al. Acrogranin, an acrosomal cysteine-rich glycoprotein, is the precursor of the growth-modulating peptides, granulins, and epithelins, and is expressed in somatic as well as male germ cells. Mol Reprod Dev. 1993;34:233–43.PubMedCrossRef Baba T, Hoff HB, Nemoto H, Lee H, Orth J, Arai Y, et al. Acrogranin, an acrosomal cysteine-rich glycoprotein, is the precursor of the growth-modulating peptides, granulins, and epithelins, and is expressed in somatic as well as male germ cells. Mol Reprod Dev. 1993;34:233–43.PubMedCrossRef
8.
Zurück zum Zitat Bateman A, Bennett HPJ. The granulin gene family: from cancer to dementia. Bioessays. 2009;31:1245–54.PubMedCrossRef Bateman A, Bennett HPJ. The granulin gene family: from cancer to dementia. Bioessays. 2009;31:1245–54.PubMedCrossRef
9.
Zurück zum Zitat Monami G, Emiliozzi V, Bitto A, Lovat F, Xu SQ, Goldoni S, et al. Proepithelin regulates prostate cancer cell biology by promoting cell growth, migration, and anchorage-independent growth. Am J Pathol. 2009;174:1037–47.PubMedPubMedCentralCrossRef Monami G, Emiliozzi V, Bitto A, Lovat F, Xu SQ, Goldoni S, et al. Proepithelin regulates prostate cancer cell biology by promoting cell growth, migration, and anchorage-independent growth. Am J Pathol. 2009;174:1037–47.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat He ZH, Bateman A. Progranulin gene expression regulates epithelial cell growth and promotes tumor growth in vivo. Cancer Res. 1999;59:3222–9.PubMed He ZH, Bateman A. Progranulin gene expression regulates epithelial cell growth and promotes tumor growth in vivo. Cancer Res. 1999;59:3222–9.PubMed
11.
Zurück zum Zitat He ZH, Ismail A, Kriazhev L, Sadvakassova G, Bateman A. Progranulin (PC-cell-derived growth factor/acrogranin) regulates invasion and cell survival. Cancer Res. 2002;62:5590–6.PubMed He ZH, Ismail A, Kriazhev L, Sadvakassova G, Bateman A. Progranulin (PC-cell-derived growth factor/acrogranin) regulates invasion and cell survival. Cancer Res. 2002;62:5590–6.PubMed
12.
Zurück zum Zitat Koo DH, Park CY, Lee ES, Ro J, Oh SW. Progranulin as a prognostic biomarker for breast cancer recurrence in patients who had hormone receptor-positive tumors: a cohort study. PLoS One. 2012;7:e39880.PubMedPubMedCentralCrossRef Koo DH, Park CY, Lee ES, Ro J, Oh SW. Progranulin as a prognostic biomarker for breast cancer recurrence in patients who had hormone receptor-positive tumors: a cohort study. PLoS One. 2012;7:e39880.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Han JJ, Yu M, Houston N, Steinberg SM, Kohn EC. Progranulin is a potential prognostic biomarker in advanced epithelial ovarian cancers. Gynecol Oncol. 2011;120:5–10.PubMedPubMedCentralCrossRef Han JJ, Yu M, Houston N, Steinberg SM, Kohn EC. Progranulin is a potential prognostic biomarker in advanced epithelial ovarian cancers. Gynecol Oncol. 2011;120:5–10.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Lu Y, Zheng L, Zhang W, Feng T, Liu J, Wang X, et al. Growth factor progranulin contributes to cervical cancer cell proliferation and transformation in vivo and in vitro. Gynecol Oncol. 2014;134:364–71.PubMedCrossRef Lu Y, Zheng L, Zhang W, Feng T, Liu J, Wang X, et al. Growth factor progranulin contributes to cervical cancer cell proliferation and transformation in vivo and in vitro. Gynecol Oncol. 2014;134:364–71.PubMedCrossRef
15.
Zurück zum Zitat Demorrow S. Progranulin: a novel regulator of gastrointestinal cancer progression. Transl Gastrointest Cancer. 2013;2:145–51.PubMedPubMedCentral Demorrow S. Progranulin: a novel regulator of gastrointestinal cancer progression. Transl Gastrointest Cancer. 2013;2:145–51.PubMedPubMedCentral
16.
Zurück zum Zitat Donald CD, Laddu A, Chandham P, Lim SD, Cohen C, Amin M, et al. Expression of progranulin and the epithelin/granulin precursor acrogranin correlates with neoplastic state in renal epithelium. Anticancer Res. 2001;21:3739–42.PubMed Donald CD, Laddu A, Chandham P, Lim SD, Cohen C, Amin M, et al. Expression of progranulin and the epithelin/granulin precursor acrogranin correlates with neoplastic state in renal epithelium. Anticancer Res. 2001;21:3739–42.PubMed
17.
Zurück zum Zitat Tang W, Lu Y, Tian QY, Zhang Y, Guo FJ, Liu GY, et al. The growth factor progranulin binds to TNF receptors and is therapeutic against inflammatory arthritis in mice. Science. 2011;332:478–84.PubMedPubMedCentralCrossRef Tang W, Lu Y, Tian QY, Zhang Y, Guo FJ, Liu GY, et al. The growth factor progranulin binds to TNF receptors and is therapeutic against inflammatory arthritis in mice. Science. 2011;332:478–84.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Yamamoto Y, Takemura M, Serrero G, Hayashi J, Yue B, Tsuboi A, et al. Increased serum GP88 (progranulin) concentrations in rheumatoid arthritis. Inflammation. 2014;37:1806–13.PubMedCrossRef Yamamoto Y, Takemura M, Serrero G, Hayashi J, Yue B, Tsuboi A, et al. Increased serum GP88 (progranulin) concentrations in rheumatoid arthritis. Inflammation. 2014;37:1806–13.PubMedCrossRef
20.
Zurück zum Zitat Zhao YP, Tian QY, Liu CJ. Progranulin deficiency exaggerates, whereas progranulin-derived Atsttrin attenuates, severity of dermatitis in mice. FEBS Lett. 2013;587:1805–10.PubMedPubMedCentralCrossRef Zhao YP, Tian QY, Liu CJ. Progranulin deficiency exaggerates, whereas progranulin-derived Atsttrin attenuates, severity of dermatitis in mice. FEBS Lett. 2013;587:1805–10.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Huang K, Chen A, Zhang X, Song Z, Xu H, Cao J, et al. Progranulin is preferentially expressed in patients with psoriasis vulgaris and protects mice from psoriasis-like skin inflammation. Immunology. 2015;145:279–87.PubMedCrossRef Huang K, Chen A, Zhang X, Song Z, Xu H, Cao J, et al. Progranulin is preferentially expressed in patients with psoriasis vulgaris and protects mice from psoriasis-like skin inflammation. Immunology. 2015;145:279–87.PubMedCrossRef
22.
Zurück zum Zitat Toh H, Chitramuthu BP, Bennett HPJ, Bateman A. Structure, function, and mechanism of progranulin; the brain and beyond. J Mol Neurosci. 2011;45:538–48.PubMedCrossRef Toh H, Chitramuthu BP, Bennett HPJ, Bateman A. Structure, function, and mechanism of progranulin; the brain and beyond. J Mol Neurosci. 2011;45:538–48.PubMedCrossRef
23.
Zurück zum Zitat Petkau TL, Leavitt BR. Progranulin in neurodegenerative disease. Trends Neurosci. 2014;37(7):388–98.PubMedCrossRef Petkau TL, Leavitt BR. Progranulin in neurodegenerative disease. Trends Neurosci. 2014;37(7):388–98.PubMedCrossRef
24.
Zurück zum Zitat Cenik B, Sephton CF, Cenik BK, Herz J, Yu G. Progranulin: a proteolytically processed protein at the crossroads of inflammation and neurodegeneration. J Biol Chem. 2012;287:32298–306.PubMedPubMedCentralCrossRef Cenik B, Sephton CF, Cenik BK, Herz J, Yu G. Progranulin: a proteolytically processed protein at the crossroads of inflammation and neurodegeneration. J Biol Chem. 2012;287:32298–306.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Naphade SB, Kigerl KA, Jakeman LB, Kostyk SK, Popovich PG, Kuret J. Progranulin expression is upregulated after spinal contusion in mice. Acta Neuropathol. 2010;119:123–33.PubMedPubMedCentralCrossRef Naphade SB, Kigerl KA, Jakeman LB, Kostyk SK, Popovich PG, Kuret J. Progranulin expression is upregulated after spinal contusion in mice. Acta Neuropathol. 2010;119:123–33.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Chen-Plotkin AS, Xiao J, Geser F, Martinez-Lage M, Grossman M, Unger T, et al. Brain progranulin expression in GRN-associated frontotemporal lobar degeneration. Acta Neuropathol. 2010;119:111–22.PubMedPubMedCentralCrossRef Chen-Plotkin AS, Xiao J, Geser F, Martinez-Lage M, Grossman M, Unger T, et al. Brain progranulin expression in GRN-associated frontotemporal lobar degeneration. Acta Neuropathol. 2010;119:111–22.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Egashira Y, Suzuki Y, Azuma Y, Takagi T, Mishiro K, Sugitani S, et al. The growth factor progranulin attenuates neuronal injury induced by cerebral ischemia-reperfusion through the suppression of neutrophil recruitment. J Neuroinflammation. 2013;10:105.PubMedPubMedCentralCrossRef Egashira Y, Suzuki Y, Azuma Y, Takagi T, Mishiro K, Sugitani S, et al. The growth factor progranulin attenuates neuronal injury induced by cerebral ischemia-reperfusion through the suppression of neutrophil recruitment. J Neuroinflammation. 2013;10:105.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Zhou M, Tang W, Fu Y, Xu X, Wang Z, Lu Y, et al. Progranulin protects against renal ischemia/reperfusion injury in mice. Kidney Int. 2015;87:918–29.PubMedCrossRef Zhou M, Tang W, Fu Y, Xu X, Wang Z, Lu Y, et al. Progranulin protects against renal ischemia/reperfusion injury in mice. Kidney Int. 2015;87:918–29.PubMedCrossRef
29.
Zurück zum Zitat Kanazawa M, Kawamura K, Takahashi T, Miura M, Tanaka Y, Koyama M, et al. Multiple therapeutic effects of progranulin on experimental acute ischaemic stroke. Brain. 2015;138:1932–48.PubMedCrossRef Kanazawa M, Kawamura K, Takahashi T, Miura M, Tanaka Y, Koyama M, et al. Multiple therapeutic effects of progranulin on experimental acute ischaemic stroke. Brain. 2015;138:1932–48.PubMedCrossRef
30.
Zurück zum Zitat Qu H, Deng H, Hu Z. Plasma progranulin concentrations are increased in patients with type 2 diabetes and obesity and correlated with insulin resistance. Mediators Inflamm. 2013;2013:360190.PubMedPubMedCentralCrossRef Qu H, Deng H, Hu Z. Plasma progranulin concentrations are increased in patients with type 2 diabetes and obesity and correlated with insulin resistance. Mediators Inflamm. 2013;2013:360190.PubMedPubMedCentralCrossRef
31.
32.
Zurück zum Zitat Zhang YY, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homolog. Nature. 1994;372:425–32.PubMedCrossRef Zhang YY, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homolog. Nature. 1994;372:425–32.PubMedCrossRef
33.
Zurück zum Zitat Magni P, Liuzzi A, Ruscica M, Dozio E, Ferrario S, Bussi I, et al. Free and bound plasma leptin in normal weight and obese men and women: relationship with body composition, resting energy expenditure, insulin-sensitivity, lipid profile and macronutrient preference. Clin Endocrinol (Oxf). 2005;62:189–96.PubMedCrossRef Magni P, Liuzzi A, Ruscica M, Dozio E, Ferrario S, Bussi I, et al. Free and bound plasma leptin in normal weight and obese men and women: relationship with body composition, resting energy expenditure, insulin-sensitivity, lipid profile and macronutrient preference. Clin Endocrinol (Oxf). 2005;62:189–96.PubMedCrossRef
34.
Zurück zum Zitat Kim HK, Shin MS, Youn BS, Namkoong C, Gil SY, Kang GM, et al. Involvement of progranulin in hypothalamic glucose sensing and feeding regulation. Endocrinology. 2011;152:4672–82.PubMedCrossRef Kim HK, Shin MS, Youn BS, Namkoong C, Gil SY, Kang GM, et al. Involvement of progranulin in hypothalamic glucose sensing and feeding regulation. Endocrinology. 2011;152:4672–82.PubMedCrossRef
35.
Zurück zum Zitat Matsubara T, Mita A, Minami K, Hosooka T, Kitazawa S, Takahashi K, et al. PGRN is a key adipokine mediating high fat diet-induced insulin resistance and obesity through IL-6 in adipose tissue. Cell Metab. 2012;15:38–50.PubMedCrossRef Matsubara T, Mita A, Minami K, Hosooka T, Kitazawa S, Takahashi K, et al. PGRN is a key adipokine mediating high fat diet-induced insulin resistance and obesity through IL-6 in adipose tissue. Cell Metab. 2012;15:38–50.PubMedCrossRef
36.
Zurück zum Zitat Youn BS, Bang SI, Kloeting N, Park JW, Lee N, Oh JE, et al. Serum progranulin concentrations may be associated with macrophage infiltration into omental adipose tissue. Diabetes. 2009;58:627–36.PubMedPubMedCentralCrossRef Youn BS, Bang SI, Kloeting N, Park JW, Lee N, Oh JE, et al. Serum progranulin concentrations may be associated with macrophage infiltration into omental adipose tissue. Diabetes. 2009;58:627–36.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Li H, Zhou B, Xu L, Liu J, Zang W, Wu S, et al. Circulating PGRN is significantly associated with systemic insulin sensitivity and autophagic activity in metabolic syndrome. Endocrinology. 2014;155:3493–507.PubMedCrossRef Li H, Zhou B, Xu L, Liu J, Zang W, Wu S, et al. Circulating PGRN is significantly associated with systemic insulin sensitivity and autophagic activity in metabolic syndrome. Endocrinology. 2014;155:3493–507.PubMedCrossRef
38.
Zurück zum Zitat Toenjes A, Fasshauer M, Kratzsch J, Stumvoll M, Blueher M. Adipokine pattern in subjects with impaired fasting glucose and impaired glucose tolerance in comparison to normal glucose tolerance and diabetes. PLoS One. 2010;5(11):e13911.CrossRef Toenjes A, Fasshauer M, Kratzsch J, Stumvoll M, Blueher M. Adipokine pattern in subjects with impaired fasting glucose and impaired glucose tolerance in comparison to normal glucose tolerance and diabetes. PLoS One. 2010;5(11):e13911.CrossRef
39.
Zurück zum Zitat Yoo H, Hwang S, Hong H, Choi H, Yang S, Choi D, et al. Implication of progranulin and C1q/TNF-Related Protein-3 (CTRP3) on inflammation and atherosclerosis in subjects with or without metabolic syndrome. PLoS One. 2013;8:e55744.PubMedPubMedCentralCrossRef Yoo H, Hwang S, Hong H, Choi H, Yang S, Choi D, et al. Implication of progranulin and C1q/TNF-Related Protein-3 (CTRP3) on inflammation and atherosclerosis in subjects with or without metabolic syndrome. PLoS One. 2013;8:e55744.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Kloeting N, Fasshauer M, Dietrich A, Kovacs P, Schoen MR, Kern M, et al. Insulin-sensitive obesity. Am J Physiol Endocrinol Metab. 2010;299:E506–15.CrossRef Kloeting N, Fasshauer M, Dietrich A, Kovacs P, Schoen MR, Kern M, et al. Insulin-sensitive obesity. Am J Physiol Endocrinol Metab. 2010;299:E506–15.CrossRef
41.
Zurück zum Zitat Hwang SL, Jeong YT, Li X, Kim YD, Lu Y, Chang YC, et al. Inhibitory cross-talk between the AMPK and ERK pathways mediates endoplasmic reticulum stress-induced insulin resistance in skeletal muscle. Br J Pharmacol. 2013;169:69–81.PubMedPubMedCentralCrossRef Hwang SL, Jeong YT, Li X, Kim YD, Lu Y, Chang YC, et al. Inhibitory cross-talk between the AMPK and ERK pathways mediates endoplasmic reticulum stress-induced insulin resistance in skeletal muscle. Br J Pharmacol. 2013;169:69–81.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Ost A, Svensson K, Ruishalme I, Brannmark C, Franck N, Krook H, et al. Attenuated mTOR signaling and enhanced autophagy in adipocytes from obese patients with type 2 diabetes. Mol Med. 2010;16:235–46.PubMedPubMedCentralCrossRef Ost A, Svensson K, Ruishalme I, Brannmark C, Franck N, Krook H, et al. Attenuated mTOR signaling and enhanced autophagy in adipocytes from obese patients with type 2 diabetes. Mol Med. 2010;16:235–46.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Liu J, Li H, Zhou B, Xu L, Kang X, Yang W, et al. PGRN induces impaired insulin sensitivity and defective autophagy in hepatic insulin resistance. Mol Endocrinol. 2015;29:528–41.PubMedCrossRef Liu J, Li H, Zhou B, Xu L, Kang X, Yang W, et al. PGRN induces impaired insulin sensitivity and defective autophagy in hepatic insulin resistance. Mol Endocrinol. 2015;29:528–41.PubMedCrossRef
44.
Zurück zum Zitat Chen X, Chang J, Deng Q, Xu J, Nguyen TA, Martens LH, et al. Progranulin does not bind tumor necrosis factor (TNF) receptors and is not a direct regulator of TNF-dependent signaling or bioactivity in immune or neuronal cells. J Neurosci. 2013;33:9202–13.PubMedPubMedCentralCrossRef Chen X, Chang J, Deng Q, Xu J, Nguyen TA, Martens LH, et al. Progranulin does not bind tumor necrosis factor (TNF) receptors and is not a direct regulator of TNF-dependent signaling or bioactivity in immune or neuronal cells. J Neurosci. 2013;33:9202–13.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Wang BC, Liu H, Talwar A, Jian J. New discovery rarely runs smooth: an update on progranulin/TNFR interactions. Protein Cell. 2015 [Epub ahead of print]. Wang BC, Liu H, Talwar A, Jian J. New discovery rarely runs smooth: an update on progranulin/TNFR interactions. Protein Cell. 2015 [Epub ahead of print].
47.
Zurück zum Zitat Toh H, Cao M, Daniels E, Bateman A. Expression of the growth factor progranulin in endothelial cells influences growth and development of blood vessels: a novel mouse model. PLoS One. 2013;8:e64989.PubMedPubMedCentralCrossRef Toh H, Cao M, Daniels E, Bateman A. Expression of the growth factor progranulin in endothelial cells influences growth and development of blood vessels: a novel mouse model. PLoS One. 2013;8:e64989.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Yang H, Cheng J, Song Z, Li X, Zhang Z, Mai Y, et al. The anti-adipogenic effect of PGRN on porcine preadipocytes involves ERK1,2 mediated PPAR gamma phosphorylation. Mol Biol Rep. 2013;40:6863–72.PubMedCrossRef Yang H, Cheng J, Song Z, Li X, Zhang Z, Mai Y, et al. The anti-adipogenic effect of PGRN on porcine preadipocytes involves ERK1,2 mediated PPAR gamma phosphorylation. Mol Biol Rep. 2013;40:6863–72.PubMedCrossRef
49.
Zurück zum Zitat Mirzaei K, Hossein-Nezhad A, Keshavarz SA, Koohdani F, Saboor-Yaraghi AA, Hosseini S, et al. Crosstalk between circulating peroxisome proliferator-activated receptor gamma, adipokines and metabolic syndrome in obese subjects. Diabetol Metab Syndr. 2013;5:79.PubMedPubMedCentralCrossRef Mirzaei K, Hossein-Nezhad A, Keshavarz SA, Koohdani F, Saboor-Yaraghi AA, Hosseini S, et al. Crosstalk between circulating peroxisome proliferator-activated receptor gamma, adipokines and metabolic syndrome in obese subjects. Diabetol Metab Syndr. 2013;5:79.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Cheng YJ, Imperatore G, Geiss LS, Wang J, Saydah SH, Cowie CC, et al. Secular changes in the age-specific prevalence of diabetes among U.S. adults: 1988–2010. Diabetes Care. 2013;36:2690–6.PubMedPubMedCentralCrossRef Cheng YJ, Imperatore G, Geiss LS, Wang J, Saydah SH, Cowie CC, et al. Secular changes in the age-specific prevalence of diabetes among U.S. adults: 1988–2010. Diabetes Care. 2013;36:2690–6.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Guariguata L. By the numbers: new estimates from the IDF Diabetes Atlas Update for 2012. Diabetes Res Clin Pract. 2012;98:524–5.PubMedCrossRef Guariguata L. By the numbers: new estimates from the IDF Diabetes Atlas Update for 2012. Diabetes Res Clin Pract. 2012;98:524–5.PubMedCrossRef
52.
Zurück zum Zitat American Diabetes Association. Classification and diagnosis of diabetes. Diabetes Care. 2015;38:S8–16.CrossRef American Diabetes Association. Classification and diagnosis of diabetes. Diabetes Care. 2015;38:S8–16.CrossRef
53.
Zurück zum Zitat InterAct Consortium, Langenberg C, Sharp SJ, Schulze MB, Rolandsson O, Overvad K, et al. Long-term risk of incident type 2 diabetes and measures of overall and regional obesity: the EPIC-InterAct case-cohort study. PLoS Med. 2012;9:e1001230.CrossRef InterAct Consortium, Langenberg C, Sharp SJ, Schulze MB, Rolandsson O, Overvad K, et al. Long-term risk of incident type 2 diabetes and measures of overall and regional obesity: the EPIC-InterAct case-cohort study. PLoS Med. 2012;9:e1001230.CrossRef
54.
Zurück zum Zitat Bi Y, Wang T, Xu M, Xu Y, Li M, Lu J, et al. Advanced research on risk factors of type 2 diabetes. Diabetes Metab Res Rev. 2012;28:32–9.PubMedCrossRef Bi Y, Wang T, Xu M, Xu Y, Li M, Lu J, et al. Advanced research on risk factors of type 2 diabetes. Diabetes Metab Res Rev. 2012;28:32–9.PubMedCrossRef
55.
Zurück zum Zitat de Souza BM, Brondani LA, Boucas AP, Sortica DA, Kramer CK, Canani LH, et al. Associations between UCP1-3826A/G, UCP2-866G/A, Ala55Val and Ins/Del, and UCP3-55C/T polymorphisms and susceptibility to type 2 diabetes mellitus: case-control study and meta-analysis. PLoS One. 2013;8:e54259.PubMedPubMedCentralCrossRef de Souza BM, Brondani LA, Boucas AP, Sortica DA, Kramer CK, Canani LH, et al. Associations between UCP1-3826A/G, UCP2-866G/A, Ala55Val and Ins/Del, and UCP3-55C/T polymorphisms and susceptibility to type 2 diabetes mellitus: case-control study and meta-analysis. PLoS One. 2013;8:e54259.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Sutton BS, Langefeld CD, Campbell JK, Haffner SM, Norris JM, Scherzinger AL, et al. Genetic mapping of a 17q chromosomal region linked to obesity phenotypes in the IRAS family study. Int J Obes (Lond). 2006;30:1433–41.CrossRef Sutton BS, Langefeld CD, Campbell JK, Haffner SM, Norris JM, Scherzinger AL, et al. Genetic mapping of a 17q chromosomal region linked to obesity phenotypes in the IRAS family study. Int J Obes (Lond). 2006;30:1433–41.CrossRef
57.
Zurück zum Zitat Fradin D, Heath S, Lathrop M, Bougneres P. Quantitative trait loci for fasting glucose in young Europeans replicate previous findings for type 2 diabetes in 2q23-24 and other locations. Diabetes. 2007;56:1742–5.PubMedCrossRef Fradin D, Heath S, Lathrop M, Bougneres P. Quantitative trait loci for fasting glucose in young Europeans replicate previous findings for type 2 diabetes in 2q23-24 and other locations. Diabetes. 2007;56:1742–5.PubMedCrossRef
58.
Zurück zum Zitat Talbert ME, Langefeld CD, Ziegler J, Mychaleckyj JC, Haffner SM, Norris JM, et al. Polymorphisms near SOCS3 are associated with obesity and glucose homeostasis traits in Hispanic Americans from the Insulin Resistance Atherosclerosis Family Study. Hum Genet. 2009;125:153–62.PubMedPubMedCentralCrossRef Talbert ME, Langefeld CD, Ziegler J, Mychaleckyj JC, Haffner SM, Norris JM, et al. Polymorphisms near SOCS3 are associated with obesity and glucose homeostasis traits in Hispanic Americans from the Insulin Resistance Atherosclerosis Family Study. Hum Genet. 2009;125:153–62.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Perusse L, Rice T, Chagnon YC, Despres JP, Lemieux S, Roy S, et al. A genome-wide scan for abdominal fat assessed by computed tomography in the Quebec Family Study. Diabetes. 2001;50:614–21.PubMedCrossRef Perusse L, Rice T, Chagnon YC, Despres JP, Lemieux S, Roy S, et al. A genome-wide scan for abdominal fat assessed by computed tomography in the Quebec Family Study. Diabetes. 2001;50:614–21.PubMedCrossRef
60.
Zurück zum Zitat Dunmore SJ, Brown JE. The role of adipokines in beta-cell failure of type 2 diabetes. J Endocrinol. 2013;216:T37–45.PubMedCrossRef Dunmore SJ, Brown JE. The role of adipokines in beta-cell failure of type 2 diabetes. J Endocrinol. 2013;216:T37–45.PubMedCrossRef
61.
Zurück zum Zitat Bergmann K, Sypniewska G. Diabetes as a complication of adipose tissue dysfunction. Is there a role for potential new biomarkers? Clin Chem Lab Med. 2013;51:177–85.PubMedCrossRef Bergmann K, Sypniewska G. Diabetes as a complication of adipose tissue dysfunction. Is there a role for potential new biomarkers? Clin Chem Lab Med. 2013;51:177–85.PubMedCrossRef
62.
Zurück zum Zitat Flehmig G, Scholz M, Kloeting N, Fasshauer M, Toenjes A, Stumvoll M, et al. Identification of adipokine clusters related to parameters of fat mass, insulin sensitivity and inflammation. PLoS One. 2014;9:e99785.PubMedPubMedCentralCrossRef Flehmig G, Scholz M, Kloeting N, Fasshauer M, Toenjes A, Stumvoll M, et al. Identification of adipokine clusters related to parameters of fat mass, insulin sensitivity and inflammation. PLoS One. 2014;9:e99785.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Blueher M, Rudich A, Kloeting N, Golan R, Henkin Y, Rubin E, et al. Two patterns of adipokine and other biomarker dynamics in a long-term weight loss intervention. Diabetes Care. 2012;35:342–9.CrossRef Blueher M, Rudich A, Kloeting N, Golan R, Henkin Y, Rubin E, et al. Two patterns of adipokine and other biomarker dynamics in a long-term weight loss intervention. Diabetes Care. 2012;35:342–9.CrossRef
64.
Zurück zum Zitat Tanaka Y, Takahashi T, Tamori Y. Circulating progranulin level is associated with visceral fat and elevated liver enzymes: significance of serum progranulin as a useful marker for liver dysfunction. Endocr J. 2014;61:1191–6.PubMedCrossRef Tanaka Y, Takahashi T, Tamori Y. Circulating progranulin level is associated with visceral fat and elevated liver enzymes: significance of serum progranulin as a useful marker for liver dysfunction. Endocr J. 2014;61:1191–6.PubMedCrossRef
65.
Zurück zum Zitat Tanaka S, Tanaka S, Iimuro S, Yamashita H, Katayama S, Akanuma Y, et al. Predicting macro- and microvascular complications in type 2 diabetes. Diabetes Care. 2013;36:1193–9.PubMedPubMedCentralCrossRef Tanaka S, Tanaka S, Iimuro S, Yamashita H, Katayama S, Akanuma Y, et al. Predicting macro- and microvascular complications in type 2 diabetes. Diabetes Care. 2013;36:1193–9.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Jha V, Garcia-Garcia G, Iseki K, Li Z, Naicker S, Plattner B, et al. Chronic kidney disease: global dimension and perspectives. Lancet. 2013;382:260–72.PubMedCrossRef Jha V, Garcia-Garcia G, Iseki K, Li Z, Naicker S, Plattner B, et al. Chronic kidney disease: global dimension and perspectives. Lancet. 2013;382:260–72.PubMedCrossRef
67.
Zurück zum Zitat Bruno RM, Gross JL. Prognostic factors in Brazilian diabetic patients starting dialysis—a 3.6-year follow-up study. J Diabetes Complicat. 2000;14:266–71.PubMedCrossRef Bruno RM, Gross JL. Prognostic factors in Brazilian diabetic patients starting dialysis—a 3.6-year follow-up study. J Diabetes Complicat. 2000;14:266–71.PubMedCrossRef
68.
Zurück zum Zitat Adler AI, Stevens RJ, Manley SE, Bilous RW, Cull CA, Holman RR, et al. Development and progression of nephropathy in type 2 diabetes: the United Kingdom Prospective Diabetes Study (UKPDS 64). Kidney Int. 2003;63:225–32.PubMedCrossRef Adler AI, Stevens RJ, Manley SE, Bilous RW, Cull CA, Holman RR, et al. Development and progression of nephropathy in type 2 diabetes: the United Kingdom Prospective Diabetes Study (UKPDS 64). Kidney Int. 2003;63:225–32.PubMedCrossRef
69.
70.
Zurück zum Zitat Gross JL, de Azevedo MJ, Silveiro SP, Canani LH, Caramori ML, Zelmanovitz T. Diabetic nephropathy: diagnosis, prevention, and treatment. Diabetes Care. 2005;28:164–76.PubMedCrossRef Gross JL, de Azevedo MJ, Silveiro SP, Canani LH, Caramori ML, Zelmanovitz T. Diabetic nephropathy: diagnosis, prevention, and treatment. Diabetes Care. 2005;28:164–76.PubMedCrossRef
71.
Zurück zum Zitat Kanasaki K, Taduri G, Koya D. Diabetic nephropathy: the role of inflammation in fibroblast activation and kidney fibrosis. Front Endocrinol (Lausanne). 2013;4:7.PubMedPubMedCentral Kanasaki K, Taduri G, Koya D. Diabetic nephropathy: the role of inflammation in fibroblast activation and kidney fibrosis. Front Endocrinol (Lausanne). 2013;4:7.PubMedPubMedCentral
72.
Zurück zum Zitat Kramer CK, Leitao CB, Pinto LC, Silveiro SP, Gross JL, Canani LH. Clinical and laboratory profile of patients with type 2 diabetes with low glomerular filtration rate and normoalbuminuria. Diabetes Care. 2007;30:1998–2000.PubMedCrossRef Kramer CK, Leitao CB, Pinto LC, Silveiro SP, Gross JL, Canani LH. Clinical and laboratory profile of patients with type 2 diabetes with low glomerular filtration rate and normoalbuminuria. Diabetes Care. 2007;30:1998–2000.PubMedCrossRef
73.
Zurück zum Zitat Afkarian M, Sachs MC, Kestenbaum B, Hirsch IB, Tuttle KR, Hinnmelfarb J, et al. Kidney disease and increased mortality risk in type 2 diabetes. J Am Soc Nephrol. 2013;24:302–8.PubMedPubMedCentralCrossRef Afkarian M, Sachs MC, Kestenbaum B, Hirsch IB, Tuttle KR, Hinnmelfarb J, et al. Kidney disease and increased mortality risk in type 2 diabetes. J Am Soc Nephrol. 2013;24:302–8.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Richter J, Focke D, Ebert T, Kovacs P, Bachmann A, Loessner U, et al. Serum levels of the adipokine progranulin depend on renal function. Diabetes Care. 2013;36:410–4.PubMedPubMedCentralCrossRef Richter J, Focke D, Ebert T, Kovacs P, Bachmann A, Loessner U, et al. Serum levels of the adipokine progranulin depend on renal function. Diabetes Care. 2013;36:410–4.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Xu L, Zhou B, Li H, Liu J, Du J, Zang W, et al. Serum levels of progranulin are closely associated with microvascular complication in type 2 diabetes. Dis Markers. 2015;2015:357279.PubMedPubMedCentralCrossRef Xu L, Zhou B, Li H, Liu J, Du J, Zang W, et al. Serum levels of progranulin are closely associated with microvascular complication in type 2 diabetes. Dis Markers. 2015;2015:357279.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Schlatzer D, Maahs DM, Chance MR, Dazard JE, Li X, Hazlett F, et al. Novel urinary protein biomarkers predicting the development of microalbuminuria and renal function decline in type 1 diabetes. Diabetes Care. 2012;35:549–55.PubMedPubMedCentralCrossRef Schlatzer D, Maahs DM, Chance MR, Dazard JE, Li X, Hazlett F, et al. Novel urinary protein biomarkers predicting the development of microalbuminuria and renal function decline in type 1 diabetes. Diabetes Care. 2012;35:549–55.PubMedPubMedCentralCrossRef
Metadaten
Titel
The role of progranulin in diabetes and kidney disease
verfasst von
Bruna Bellincanta Nicoletto
Luis Henrique Canani
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Diabetology & Metabolic Syndrome / Ausgabe 1/2015
Elektronische ISSN: 1758-5996
DOI
https://doi.org/10.1186/s13098-015-0112-6

Weitere Artikel der Ausgabe 1/2015

Diabetology & Metabolic Syndrome 1/2015 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.