Loading

PTEN-Induced Kinase 1 Regulates Mitochondrial Integrity and Insulin Secretion in Mouse Pancreatic ? –Cells

Research Article | Open Access | Volume 1 | Issue 1

  • 0. Both authors contribute equally
  • 1. Department of Internal Medicine, Division of Endocrinology and Molecular Medicine, University of Kentucky, USA
  • 2. Department of Anatomy and Neurobiology, University of Kentucky, USA
  • 3. Graduate Center for Nutritional Sciences, University of Kentucky, USA
+ Show More - Show Less
Corresponding Authors
Preetha Shridas, Department of Internal Medicine, Division of Endocrinology and Molecular Medicine, University of Kentucky, Lexington, KY 40536, USA, Tel: 859-323-4933. Hansruedi Büeler, Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY 40536, USA, Tel: 859-323-0307.
Abstract

PTEN-induced kinase 1 (PINK1) is linked to recessive familial Parkinson’s disease (PD). PINK1 and Parkin (a PD-linked ubiquitin ligase) regulate mitochondrial quality control by promoting the degradation of depolarized mitochondria via mitophagy. In addition, PINK1 stimulates macro-autophagy (autophagy) by binding to Beclin-1. Mice and cells lacking PINK1 develop multiple mitochondrial defects that may be mediated by oxidative stress and abnormal mitophagy. Mitochondrial dysfunction is also implicated in the pathogenesis of type-2 diabetes (T2D), and several etiologic studies have shown a link between T2D and PD. Here we investigated the effects of PINK1 ablation on mitochondrial morphology and autophagy in mouse pancreatic β-cells. In addition, we studied how PINK1 deficiency and over-expression affects glucosestimulated insulin secretion (GSIS) from pancreatic islets and MIN6 pancreatic β-cells. We show that loss of PINK1 in mice leads to mitochondrial enlargement/swelling in β-cells accompanied by an accumulation of autophagosomes and ubiquitinated protein inclusions. PINK1 deficiency impairs GSIS but not insulin production in pancreatic islets, while over-expression of PINK1 in MIN6 β-cells increases GSIS. To our knowledge, this is the first demonstration that a familial PD-linked protein regulates mitochondrial integrity and GSIS in pancreatic β-cells. Our results provide a possible mechanistic explanation for the link between T2D and PD and implicate PINK1 as a risk factor in the development of diabetes. Through its ability to regulate mitochondrial quality control and GSIS in pancreatic β-cells PINK1 may be explored as a novel target for the treatment of T2D.

Keywords

•    PINK1
•    Parkinson’s disease
•    Diabetes
•    Insulin
•    Mitochondria
•    Autophagy
•    Pancreas

Citation

Shridas P, Zhi L, Akundi RS, Webb NR, Pearson KJ, et al. (2013) PTEN-Induced Kinase 1 Regulates Mitochondrial Integrity and Insulin Secretion in Mouse Pancreatic β –Cells. J Endocrinol Diabetes Obes 1(1): 1007.

INTRODUCTION

Mutations in PINK1 cause early-onset recessive Parkinson’s disease (PD) [1-3]. PINK1 is a serine/threonine kinase with an N-terminal mitochondrial signal sequence [1]. Full-length PINK1 localizes to mitochondria, while an N-terminally cleaved form is present in the cytoplasm [4-6]. Consistent with this, PINK1 regulates both mitochondrial and cytosolic pathways to protect neurons against cell death [7-9]. PINK1 and Parkin cooperate to promote the selective degradation of depolarized mitochondria through mitophagy [10-12], a mechanism central to mitochondrial quality control. PINK1 also stimulates general autophagy by interacting with Beclin-1, which is independent of the PINK1 kinase activity [13]. In addition, PINK1 phosphorylates the mitochondrial chaperone TRAP1, thereby protecting neurons against oxidative stress-induced apoptosis [8]. PINK1-deficient mice show age-dependent dopamine loss, impaired dopamine release and various abnormalities of mitochondrial function [14-16]. PINK1-deficient primary cells show respiratory defects and accumulation of enlarged and swollen mitochondria, possibly due to increased fusion of non-degraded, depolarized mitochondria with the normal mitochondrial network [17,18]. Finally, PINK1 protects cells against stress-induced apoptosis through enhancing signal transduction by the cell survival kinase Akt [9-19,20]. Although mutations in PINK1 have only been linked to PD, mitochondrial metabolism and autophagy are important to maintain homeostasis of cells and organs in general. In particular, pancreatic β-cell function relies heavily on mitochondrial bioenergetics, and disturbances in mitochondrial function, autophagy and oxidative stress are implicated in the pathogenesis of diabetes [21-24]. Interestingly, several studies suggest that T2D is a risk factor for the development of PD [25,26], but the mechanistic links are poorly understood. Here, we investigated the effects of PINK1 ablation and over-expression on glucose-stimulated insulin secretion (GSIS) from pancreatic islets and cultured mouse MIN6 pancreatic β-cells. We show that loss of PINK1 impairs GSIS from pancreatic islets. In contrast, over-expression of PINK1 in MIN6 cells increases GSIS. In addition, we demonstrate that PINK1-deficient pancreatic β-cells (in sections of the pancreas) harbor significantly enlarged and swollen mitochondria and display accumulation of autophagosomes and ubiquitin-positive protein inclusions. Our results show that loss of PINK1 results in impaired GSIS from pancreatic β-cells and that this defect is associated with abnormal mitochondrial integrity and possibly autophagy. Mutations in PINK1 may increase the risk for diabetes and PINK1 may be explored as a novel target for the treatment of T2D.

MATERIALS AND METHODS

Animals

PINK1-deficient mice used in this study have been described elsewhere [14]. All work has been conducted according to national and international guidelines and has been approved by the Animal Care and Use Committee of the University of Kentucky. WT and PINK1-deficient littermates were analyzed at 4-5 months of age unless stated otherwise. Mice were housed under 12-hour light-dark cycles and had free access to food (Harlan #2918) and water.

Analysis of PINK1 expression

Total RNA from 150-200 wildtype C57BL/6 mouse islets and from MIN6 cells was isolated and 0.5 µg RNA was converted to cDNA (High Capacity cDNA Reverse Transcription kit, Applied Biosystems). Two µl cDNA reaction was used for PCR with intron-spanning primers complementary to exon-1 and exon-3 of the Pink1 gene (35 cycles of 94?C/30s; 58?C/45sec and 72?C /1 min). As a negative control the cDNA synthesis step (reverse transcriptase) was omitted. PINK1 protein was detected in 5 µm-thick paraffin sections of pancreata that were fixed in 4% paraformaldehyde at 4?C overnight. For immunohistochemistry, sections were de-waxed, incubated in citrate buffer (10mM, pH 6.0) for antigen retrieval in a preheated steamer for 20 min, blocked for 1 hour in blocking buffer (10% normal donkey serum, 0.2% Triton X-100, and 0.1% sodium azide in TBS) and stained in blocking buffer at 4?C overnight with rabbit anti-PINK1 antibody (1:100, NBP1-49678, Novus) and goat anti-insulin antibody (1:500, sc-7839, Santa Cruz). After washing sections were incubated for 1 hour in blocking buffer without Triton X-100 with donkey anti-rabbit IgG-Alexa 488 and donkey anti-goat IgG-Alexa 594 (both at 1:300 dilution, from Invitrogen). Sections were mounted in ProLong Gold medium (Invitrogen) and immediately imaged using a Leica TCS SP5 AOBS inverted confocal microscope at an emission wavelength of 488nm and 594nm.

Isolation of islets and GSIS

Mouse islets were isolated via intraductal collagenase (Roche) digestion and Histopaque gradient (Histopaque-1077, Sigma) centrifugation by a method adopted by the Islet Procurement and Analysis Core, Diabetes Research and Training Center, Vanderbilt University Medical Center (Nashville, TN). Briefly, pancreata from mice were infused with 5ml of 0.5mg/ml collagenase in Hank’s balanced saltsolution (HBSS), surgically removed and collagenase digestion was continued for 10-15 min at 37?C and then stopped by addition of chilled HBSS containing 10% FBS. Islets were purified using a Histopaque gradient centrifugation step, individually picked by hand and cultured overnight in RPMI medium containing 5mM glucose, 10% (v/v) FBS, 100 U/ ml penicillin and 100µg/ml streptomycin. For GSIS islets were transferred to culture inserts (Greiner Bio-One) in 12-well plates (25 islets per well) and equilibrated for 1 hour in buffer 1 (DMEM, 38mM sodium bicarbonate, 4mM L-glutamine, 1mM sodium pyruvate, 4.65mM HEPES and 1 g/l BSA) supplemented with 5mM (low) glucose. Basal insulin secretion was measured after 40 min incubation in buffer 1 supplemented with 5mM glucose. Thereafter the medium was replaced with buffer 1 containing 20mM (high) glucose and the insulin content in the medium measured again after 40 min. Total intracellular insulin was determined after disrupting the cells with acid-ethanol (75% ethanol, 0.2M HCl). Percent of secreted insulin was calculated as the ratio of secreted to intracellular insulin. All insulin measurements were done with the ultra-sensitive mouse insulin ELISA kit (Crystal Chem Inc.) according to the manufacturer’s instructions.

GSIS with transfected MIN6 cells

MIN6 cells were cultured in DMEM, 15% heat-inactivated FBS, 2mM L-glutamine, 45mM β- mercaptoethanol, 100 U/ml penicillin and 100 µg/ml streptomycin. MIN6 cells (passage number <30, 75-80% confluent) were transfected with 1µg pCMVTNT PINK1 C-myc plasmid (Addgene plasmid 13314, deposited by Mark Cookson) or pEGFP-N1 (Clontech) using Lipofectamine 2000 (Invitrogen). Medium was changed 24 hours after transfection. 48 hours after transfection cells were washed once with Kreb’s Ringer buffer containing 0.2% BSA (KRB-BSA) and 5mM glucose and then equilibrated in the same buffer for 1 hour. The medium was replaced with fresh KRB-BSA containing either 5mM (low) glucose or 20mM (high) glucose for 40 min. Insulin concentrations in medium were determined as above and normalized to total cell protein (measured with the BCA kit, Pierce).

Analysis of mitochondrial morphology (electron microscopy)

Mice were euthanized by CO2 inhalation. Pancreata were dissected immediately, rinsed with 0.1 M cacodylate buffer (pH 7.4) and immersed in freshly made 4% paraformaldehyde/3.5% glutaraldehyde/0.1M cacodylate buffer (pH 7.4) (Electron Microscopy Sciences). Pancreata were cut into small pieces (~1 mm3 ) and kept in fixation solution for 2 hours at 4?C. After four washes in 0.1M cacodylate buffer containing 5% sucrose (15 min each), tissue pieces were post-fixed in 1% osmium tetroxide for 90 min at 4?C, dehydrated in graded ethanol (from 50%-100% 10 min at 4?C). Further dehydration was achieved by two 15-min incubations in absolute ethanol and two 15-min incubations in propylene oxide (PO) at RT. Tissues were infiltrated with 50% resin (tEpon)/50% propylene oxide with accelerator overnight at RT under a 60-watt lamp. The PO/resin mixture was poured off and tissues placed twice for 1 hour in fresh 100% resin. Tissues were embedded in BEEM capsules (Ted Pella Inc., CA) and allowed to polymerize at 60?C for 48 hrs. The beam capsules were snapped off and ultrathin sections of pancreata were prepared in the University of Kentucky Imaging Facility and imaged using a Philips Tecnai Biotwin-12 transmission electron microscope at the magnifications indicated in the figure legend. Mitochondrial size was quantified using NIH image J software (http://imagej. nih.gov/ij).

Pancreas immunohistochemistry for LC3 and Ubiquitin

Procedures were the same as described above for detection of PINK1 expression. Antibodies used were rabbit anti-LC3 (1:50, Abcam ab58610) and rabbit anti-ubiquitin (1:100, Santa Cruz sc-9133). The anti- LC3 antibody recognizes both LC3I and LC3II isoforms, but LC3II is specific for autophagosomes and in IHC thus appears as punctae in cells.

Data analysis

Data were analyzed by ANOVA and two-tailed t-test. Values of p<0.05 were considered statistically significant.

RESULTS

PINK1 deficiency impairs GSIS in pancreatic mouse islets

Mitochondria play an important function in the regulation of both neurotransmission [27] and insulin secretion [21- 28]. Based on our previous studies showing mitochondrial defects in vivo and in several primary cell types of PINK1-deficient mice [14,17] we hypothesized that loss of PINK1 may also affect mitochondria in pancreatic β-cells, leading to impairments in GSIS. Using reverse-transcription PCR we first demonstrated that PINK1 mRNA is expressed in pancreatic islets and MIN6 cells, a mouse pancreatic β-cell line (Figure 1A).

PINK1 is expressed in pancreatic islet cells and MIN6 cells. (A) Expression of PINK1 mRNA in mouse pancreatic islets and the MIN6  pancreatic ?-cell line. Total RNA was isolated and expression of Pink1 mRNA was  analyzed as described in Materials and Methods. (B) Expression of PINK1 protein in ?-cells of the mouse pancreas. Paraffin  sections of normal mouse pancreas were stained with antibodies against PINK1  (green) and insulin (red) and imaged by confocal microscopy as described in  Materials and Methods. Scale bar, 20 µm

Figure 1 PINK1 is expressed in pancreatic islet cells and MIN6 cells. (A) Expression of PINK1 mRNA in mouse pancreatic islets and the MIN6 pancreatic β-cell line. Total RNA was isolated and expression of Pink1 mRNA was analyzed as described in Materials and Methods. (B) Expression of PINK1 protein in β-cells of the mouse pancreas. Paraffin sections of normal mouse pancreas were stained with antibodies against PINK1 (green) and insulin (red) and imaged by confocal microscopy as described in Materials and Methods. Scale bar, 20 µm.

In addition, we showed that PINK protein is detectable in pancreatic β- cells by co-localization with insulin in sections of paraffin-embedded pancreas (Figure 1B). To test the hypothesis that PINK1 is important for β-cell function, we isolated primary islets from WT and PINK1-/- mice and performed GSIS assays. There was no significant difference in basal (5 mM glucose) insulin secretion between islets from WT and PINK1-/- mice and high (20 mM) glucose led to a significant increase in insulin release from islets of both genotypes (Figure 2).

Loss of PINK1 reduces glucose-stimulated insulin secretion in  pancreatic ?-cells: Insulin secretion from isolated pancreatic islets of 4-5 month-old WT and  PINK1-/- mice (25 islets per animal). Islets were cultured overnight, incubated  first for 40 min in buffer containing 5mM (low) glucose and then transferred to  buffer with 20mM (high) glucose for 40 min. Insulin content in low-glucose and  high-glucose medium was measured at the end of the 40 min incubation times.  Secretion of insulin expressed as percentage of total cellular insulin (determined  in acid ethanol-disrupted islets as described in Methods). Mean ± SEM, **p<0.01.  Experiments were repeated two different times with similar results.

Figure 2 Loss of PINK1 reduces glucose-stimulated insulin secretion in pancreatic β-cells: Insulin secretion from isolated pancreatic islets of 4-5 month-old WT and PINK1-/- mice (25 islets per animal). Islets were cultured overnight, incubated first for 40 min in buffer containing 5mM (low) glucose and then transferred to buffer with 20mM (high) glucose for 40 min. Insulin content in low-glucose and high-glucose medium was measured at the end of the 40 min incubation times. Secretion of insulin expressed as percentage of total cellular insulin (determined in acid ethanol-disrupted islets as described in Methods). Mean ± SEM, **p<0.01. Experiments were repeated two different times with similar results.

However, GSIS was significantly lower in islets isolated from PINK1-/- mice compared to WT islets (Figure 2, 60% reduction), Thus, PINK1 is required for maximum GSIS in mouse pancreatic islets.

PINK1 activates GSIS in MIN6 cells

To further investigate the role of PINK1 in GSIS, we utilized a gain-of-function approach where we measured GSIS in MIN6 cells transfected with either a plasmid carrying the PINK1 cDNA (MIN6- PINK1) or an EGFP control vector (MIN6-EGFP). Transfection efficiency was estimated at 40% (Figure 3A),

PINK1 enhances glucose-stimulated insulin secretion in MIN6  cells: MIN6 cells were a gift from Dr. Sabire Ozcan (University of Kentucky).  Cells were kept at passage number <30 and transfected when 75-80% confluent  as described in Materials and Methods. (A) MIN6 cells transfected with EGFP  expression plasmid, showing EGFP fluorescence (top) and bright field picture  (bottom) of the same view field. (B) Basal and glucose-stimulated insulin  secretion. GSIS assay was performed in MIN6 cells 48 hours after transfection  as described in Materials and Methods (n=4-6 wells for each MIN6-EGFP and  MIN6-PINK1). The amount of secreted insulin was normalized to total cellular  protein. Mean ± SEM, *p<0.05. Experiments were repeated two different times  with similar results.

Figure 3 PINK1 enhances glucose-stimulated insulin secretion in MIN6 cells: MIN6 cells were a gift from Dr. Sabire Ozcan (University of Kentucky). Cells were kept at passage number <30 and transfected when 75-80% confluent as described in Materials and Methods. (A) MIN6 cells transfected with EGFP expression plasmid, showing EGFP fluorescence (top) and bright field picture (bottom) of the same view field. (B) Basal and glucose-stimulated insulin secretion. GSIS assay was performed in MIN6 cells 48 hours after transfection as described in Materials and Methods (n=4-6 wells for each MIN6-EGFP and MIN6-PINK1). The amount of secreted insulin was normalized to total cellular protein. Mean ± SEM, *p<0.05. Experiments were repeated two different times with similar results.

and MIN6-PINK1 cells expressed ~4-fold higher levels of PINK1 mRNA compared to MIN6-EGFP cells as determined by quantitative real-time PCR (data not shown). Basal insulin secretion in MIN6- PINK1 and MIN6-EGFP cells was the same (Figure 3B, 5 mM glucose). Interestingly, over-expression of PINK1 increased GSIS to a level significantly higher than in MIN6-EGFP cells (Figure 3B, 20 mM glucose). Combined with the data shown above, these results suggest that PINK1 expression levels positively correlate with GSIS from pancreatic β-cells.

Loss of PINK1 leads to mitochondrial swelling and cristae defects in pancreatic β –cells

To study if PINK1 deficiency affects mitochondrial integrity in pancreatic β-cells, we analyzed mitochondrial morphology in pancreatic islets of WT and PINK1-/- mice using electron microscopy. Consistent with results in other cells [17] we found that the average size of mitochondria in β-cells from PINK1-/- mice is increased (Figure 4),

Mitochondrial swelling in PINK1-deficient pancreatic ? –cells:  Pancreata were embedded, processed and cut for analysis by electron microscopy  as described in Materials and Methods. (A-F) Electron microscopy images of  mitochondria in pancreatic ?-cells of 4-month and 8-month old WT (A,C,E) and  PINK1-/- (B,D,F) mice (fold magnification indicated). (G) Statistical analysis of  mitochondrial size. Thirty-seven to sixty-seven randomly chosen mitochondria  in pancreatic ?-cells were analyzed at 49,000x magnification using NIH Image J  software for n=3 mice per genotype and the means were compared by t-test. WT  mice, black bars; PINK1-/- mice, grey bars; mean ± SD; **p<0.01). In panels B, D  and F arrows point to enlarged/swollen mitochondria with cristae fragmentation.  Asterisks in panels A- D indicate insulin granules. Scale bars, 0.5 µm

Figure 4 Mitochondrial swelling in PINK1-deficient pancreatic β –cells: Pancreata were embedded, processed and cut for analysis by electron microscopy as described in Materials and Methods. (A-F) Electron microscopy images of mitochondria in pancreatic β-cells of 4-month and 8-month old WT (A,C,E) and PINK1-/- (B,D,F) mice (fold magnification indicated). (G) Statistical analysis of mitochondrial size. Thirty-seven to sixty-seven randomly chosen mitochondria in pancreatic β-cells were analyzed at 49,000x magnification using NIH Image J software for n=3 mice per genotype and the means were compared by t-test. WT mice, black bars; PINK1-/- mice, grey bars; mean ± SD; **p<0.01). In panels B, D and F arrows point to enlarged/swollen mitochondria with cristae fragmentation. Asterisks in panels A- D indicate insulin granules. Scale bars, 0.5 µm

with occasional accumulation of severely swollen mitochondria (arrows in Figure 4B, D and F). In addition, in many PINK1-/- mitochondria the cristae (folds of the inner membrane carrying the respiratory complexes) show reduced density and signs of fragmentation (arrows in Figure 4B and 4D). In contrast, the number of insulin granules was similar in WT and PINK1-/- β-cells (Figure 4E-F), in agreement with the normal intracellular insulin content and basal insulin secretion of PINK1-/- islets. Mitochondrial enlargement in PINK1-/- β-cells is present at 4 months (Figure 4B and 4G) and persists at 8 months (Figure 4D and 4F). These data show that loss of PINK1 leads to mitochondrial dysfunction in pancreatic β-cells.

PINK1-deficient pancreatic β -cells display accumulation of autophagosomes and ubiquitin-positive inclusions

In addition to promoting mitophagy of depolarized mitochondria PINK1 interacts with Beclin-1 to stimulate macroautophagy (autophagy) [13]. In pancreatic β-cells autophagy is important for the degradation of ubiquitinated protein aggregates that form as a result of diabetes-induced oxidative stress [24]. To study if the loss of PINK1 alters β-cell autophagy we stained sections of the pancreas with an antibody against LC3, which when incorporated into autophagosomes (LC3II isoform) appears as punctae in cells. Compared to WT mice, pancreatic β-cells from PINK1-deficient mice contained significantly more punctae/autophagosomes (Figure 5A).

Increased numbers of autophagosomes and ubiquitin-positive  inclusions in pancreatic ? -cells of PINK1-/- mice: Paraffin sections of  pancreata were stained with antibodies against (A) LC3 and insulin to detect autophagosomes and (B) ubiquitin and insulin  to detect ubiquitinated inclusions in ?-ells. Panels show overlays of green  (Alexa488) and red (Alexa594) signals captured by confocal microscopy and are  representative of three animals per genotype

Figure 5 Increased numbers of autophagosomes and ubiquitin-positive inclusions in pancreatic β -cells of PINK1-/- mice: Paraffin sections of pancreata were stained with antibodies against (A) LC3 and insulin to detect autophagosomes and (B) ubiquitin and insulin to detect ubiquitinated inclusions in β-ells. Panels show overlays of green (Alexa488) and red (Alexa594) signals captured by confocal microscopy and are representative of three animals per genotype.

Moreover, PINK1-/- β-cells accumulate ubiquitin-positive inclusions (Figure 5B). While increased numbers of autophagosomes seem inconsistent with the stimulatory role of PINK1 in autophagy induction [13], it should be noted that compensatory up-regulation of autophagy has been observed in PINK1-deficient neurons [29]. Thus, the higher abundance of ubiquitin- positive inclusions may be due to reduced clearance of autophagosomes (fusion with lysosomes) rather than decreased autophagy induction. However, further studies in the future are necessary to substantiate a direct role of PINK1 in the regulation of β-cell autophagy.

DISCUSSION

Clinical studies suggest a link between T2D and PD, especially in younger patients [25,26]. Both PD and diabetes are characterized by mitochondrial dysfunction [21,30] and several genes linked to early-onset familial PD regulate mitochondrial function and quality control [30,31]. However, to date little is known about how mutations in familial PD genes affect the function and homeostasis of tissues implicated in the development of T2D. Here, we studied the effects of PINK1 deficiency on mitochondrial integrity, autophagy and insulin secretion in pancreatic β-cells. We show that PINK1 deficiency results in a significant decrease in GSIS from pancreatic islets without affecting intracellular insulin levels. The insulin secretion defect is associated with abnormally swollen mitochondria in PINK1-deficient β-cells in vivo, many of which develop cristae fragmentation. We recently showed that similarly enlarged mitochondria with disintegrated cristae are present in primary neurons of PINK1-deficient mice [17]. Results presented here demonstrate that mitochondrial quality control exerted by PINK1 is also important in β-cells. Moreover, we found that loss of PINK1 leads to an increased number of LC3II-positive autophagosomes and accumulation of ubiquitinated protein inclusions, suggesting altered autophagy in PINK1-/- β-cells. However, our experiments do not proof a direct involvement of PINK1 in the regulation of β-cell autophagy. It is possible that loss of PINK1 results in compensatory up-regulation of autophagy secondary to other defects (e.g., oxidative stress), as has been shown in cultured neuronal cells [29]. Future studies investigating whether PINK1 binds to Beclin-1 in mouse β-cells and/or if accumulation of autophagosomes is due to reduced autophagic flux are necessary to conclude that PINK1 directly controls autophagy in pancreatic β-cells. Nonetheless, it is interesting that several phenotypes of PINK1-/- β-cells are also observed in states of diabetes. For example, diabetes-induced oxidative stress in pancreatic β- cells leads to ubiquitination and storage of proteins into cytoplasmic aggregates, the number of which increases upon autophagy inhibition [24]. In addition, β-cell specific ablation of the autophagic machinery results in progressive accumulation of ubiquitinated proteins and swollen mitochondria as well as reduced glucose-stimulated insulin secretion [32,33]. Finally, long-term exposure to fatty acids and glucose blocks autophagic flux in β-cells, leading to the accumulation of autophagosomes, impaired autophagic turnover of long-lived proteins and reduced insulin secretion [34]. The autophagy inducer rapamycin ameliorated defects in autophagic flux and insulin release, showing that impaired autophagy contributes to insulin secretion defects [34]. Taken together, these results demonstrate the critical importance of mitochondria and autophagy for GSIS and suggest that mutations in PINK1 may be a risk factor for diabetes by causing abnormalities in mitochondrial integrity and autophagy in β-cells similar to those induced by high-calorie diet and obesity. It has recently been shown that DJ-1, which acts in parallel with PINK1 and Parkin to control mitochondrial function and autophagy [35], is a target for oxidation in MIN6 cells treated with high glucose [36]. Moreover, DJ-1 over-expression protected MIN6 cells from oxidative stress-induced cell death and impairments in insulin secretion [36]. Like PINK1, over-expression of DJ-1 increased GSIS from MIN6 cells [36]. Finally, over-expression of α-Synuclein (αS), which is linked to sporadic and dominant familial PD, reduced insulin secretion from MIN6 cells and mouse islets, albeit only at low glucose concentrations [37]. In contrast, islets from αS knockout mice showed elevated insulin secretion [37]. As a mechanism, it has been suggested that αS binds to insulin granules and the KATP channel, acting as an inhibitor of insulin secretion [37]. However, αS also inhibits mitochondrial complex I activity and autophagy [38,39], which if it occurred in β-cells may contribute to impaired insulin release. Finally, it has been shown that mice lacking insulin-degrading enzyme (Ide knockout mice) have impaired GSIS and autophagic flux [40]. Ide knockout mice showed accumulation of αS in pancreatic β-cells and over-expression of αS alone was sufficient to reduce GSIS [40].

CONCLUSIONS

The results presented in this work and previous studies implicating DJ-1 and αS in islet cell function and stress resistance support the notion that mutations in familial PD genes may be risk factors for T2D and provide a potential explanation for the known association between T2D and PD. Abnormal mitochondrial function and autophagy could be shared pathogenic mechanisms, because these processes are affected by all PD genes discussed above (PINK1, DJ-1, αS) and are also compromised in sporadic PD [30,41,42]. Consequently, bestowing β-cells with an enhanced capacity for mitochondrial quality control and stress resistance through over-expression or activation of recessive PD genes [8,10,11,36] may lead to improved treatments for T2D in the future. The fact that over-expression of both PINK1 (shown here) and DJ-1 [36] increases GSIS in MIN6 cells lends further support to this idea. However, autophagy needs to be well balanced because both reduced and excessive autophagy can be detrimental to β-cell function. Defects other than those investigated here may exist and contribute to the phenotype of reduced GSIS in PINK1-deficient islets, most notably increased oxidative stress [8,29,43]. In addition, lack of PINK1 may affect glucose-dependent intracellular signaling through effects on Akt activity [9,19]. Finally, because all our studies have been performed with isolated islets future experiments with animals are necessary to determine if reduced insulin secretion leads to a diabetic status in vivo. This may depend on age, genetic background and how loss of PINK1 affects the physiology of other organs involved in glucose homeostasis, including skeletal muscle and liver.

ACKNOWLEDGMENTS

We thank Sabire Ozcan for MIN6 cells and Mary Gail Engle and James Begley from the University of Kentucky Imaging Facility for excellent technical help with EM studies.

FUNDING ACKNOWLEDGEMENT

This work was supported in part by the following funding agencies: Parkinson Schweiz to HB, NIH/NIGMS Institutional Development Award supporting PS and KJP (8 P20 GM103527- 05), NIH to NRW (R01 DK082419). The authors have no financial or other conflict of interests to disclose.

REFERENCES

1. Valente EM, Abou-Sleiman PM, Caputo V, Muqit MM, Harvey K, Gispert S, et al. Hereditary early-onset Parkinson’s disease caused by mutations in PINK1. Science. 2004; 304: 1158-1160.

2. Hatano Y, Li Y, Sato K, Asakawa S, Yamamura Y, Tomiyama H, et al. Novel PINK1 mutations in early-onset parkinsonism. Ann Neurol. 2004; 56: 424-427.

3. Bonifati V, Rohé CF, Breedveld GJ, Fabrizio E, De Mari M, Tassorelli C, et al. Early-onset parkinsonism associated with PINK1 mutations: frequency, genotypes, and phenotypes. Neurology. 2005; 65: 87-95.

4. Beilina A, Van Der Brug M, Ahmad R, Kesavapany S, Miller DW, Petsko GA, et al. Mutations in PTEN-induced putative kinase 1 associated with recessive parkinsonism have differential effects on protein stability. Proc Natl Acad Sci U S A. 2005; 102: 5703-5708.

5. Lin W, Kang UJ. Characterization of PINK1 processing, stability, and subcellular localization. J Neurochem. 2008; 106: 464-474.

6. Takatori S, Ito G, Iwatsubo T. Cytoplasmic localization and proteasomal degradation of N-terminally cleaved form of PINK1. Neurosci Lett. 2008; 430: 13-17.

7. Haque ME, Thomas KJ, D’Souza C, Callaghan S, Kitada T, Slack RS, et al. Cytoplasmic Pink1 activity protects neurons from dopaminergic neurotoxin MPTP. Proc Natl Acad Sci U S A. 2008; 105: 1716-1721.

8. Pridgeon JW, Olzmann JA, Chin LS, Li L. PINK1 protects against oxidative stress by phosphorylating mitochondrial chaperone TRAP1. PLoS Biol. 2007; 5: e172.

9. Murata H, Sakaguchi M, Jin Y, Sakaguchi Y, Futami J, Yamada H, et al. A new cytosolic pathway from a Parkinson disease-associated kinase, BRPK/PINK1: activation of AKT via mTORC2. J Biol Chem. 2011; 286: 7182-7189.

10. Narendra DP, Jin SM, Tanaka A, Suen DF, Gautier CA, Shen J, et al. PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol. 2010; 8: e1000298.

11. Vives-Bauza C, Zhou C, Huang Y, Cui M, de Vries RL, Kim J, et al. PINK1- dependent recruitment of Parkin to mitochondria in mitophagy. Proc Natl Acad Sci U S A. 2010; 107: 378-383.

12. Matsuda N, Sato S, Shiba K, Okatsu K, Saisho K, Gautier CA, et al. PINK1 stabilized by mitochondrial depolarization recruits Parkin to damaged mitochondria and activates latent Parkin for mitophagy. J Cell Biol. 2010; 189: 211-221.

13. Michiorri S, Gelmetti V, Giarda E, Lombardi F, Romano F, Marongiu R, et al. The Parkinson-associated protein PINK1 interacts with Beclin1 and promotes autophagy. Cell Death Differ. 2010; 17: 962-974.

14. Akundi RS, Huang Z, Eason J, Pandya JD, Zhi L, Cass WA, et al. Increased mitochondrial calcium sensitivity and abnormal expression of innate immunity genes precede dopaminergic defects in Pink1-deficient mice. PLoS One. 2011; 6: e16038.

15. Kitada T, Pisani A, Porter DR, Yamaguchi H, Tscherter A, Martella G, et al. Impaired dopamine release and synaptic plasticity in the striatum of PINK1-deficient mice. Proc Natl Acad Sci U S A. 2007; 104: 11441- 11446.

16. Gautier CA, Kitada T, Shen J. Loss of PINK1 causes mitochondrial functional defects and increased sensitivity to oxidative stress. Proc Natl Acad Sci U S A. 2008; 105: 11364-11369.

17. Akundi RS, Zhi L, Sullivan PG, Büeler H. Shared and Cell Type-Specific Mitochondrial Defects and Metabolic Adaptations in Primary Cells from PINK1-Deficient Mice. Neurodegener Dis. 2012; .

18. Heeman B, Van den Haute C, Aelvoet SA, Valsecchi F, Rodenburg RJ, Reumers V, et al. Depletion of PINK1 affects mitochondrial metabolism, calcium homeostasis and energy maintenance. J Cell Sci. 2011; 124: 1115-1125

19. Akundi RS, Zhi L, Büeler H. PINK1 enhances insulin-like growth factor-1-dependent Akt signaling and protection against apoptosis. Neurobiol Dis. 2012; 45: 469-478.

20. Sánchez-Mora RM, Arboleda H, Arboleda G. PINK1 overexpression protects against C2-ceramide-induced CAD cell death through the PI3K/AKT pathway. J Mol Neurosci. 2012; 47: 582-594.

21. Supale S, Li N, Brun T, Maechler P. Mitochondrial dysfunction in pancreatic β cells. Trends Endocrinol Metab. 2012; 23: 477-487.

22. Ma ZA, Zhao Z, Turk J. Mitochondrial dysfunction and β-cell failure in type 2 diabetes mellitus. Exp Diabetes Res. 2012; 2012: 703538.

23. Quan W, Lim YM, Lee MS. Role of autophagy in diabetes and endoplasmic reticulum stress of pancreatic β-cells. Exp Mol Med. 2012; 44: 81-88.

24. Kaniuk NA, Kiraly M, Bates H, Vranic M, Volchuk A, Brumell JH. et al. Ubiquitinated-protein aggregates form in pancreatic beta-cells during diabetes-induced oxidative stress and are regulated by autophagy. Diabetes. 2007; 56: 930-939.

25. Hu G, Jousilahti P, Bidel S, Antikainen R, Tuomilehto J. Type 2 diabetes and the risk of Parkinson’s disease. Diabetes Care. 2007; 30: 842-847.

26. Schernhammer E, Hansen J, Rugbjerg K, Wermuth L, Ritz B. Diabetes and the risk of developing Parkinson’s disease in Denmark. Diabetes Care. 2011; 34: 1102-1108.

27. Vos M, Lauwers E, Verstreken P. Synaptic mitochondria in synaptic transmission and organization of vesicle pools in health and disease. Front Synaptic Neurosci. 2010; 2: 139.

28. Wiederkehr A, Wollheim CB. Mitochondrial signals drive insulin secretion in the pancreatic β-cell. Mol Cell Endocrinol. 2012; 353: 128-137.

29. Dagda RK, Cherra SJ 3rd, Kulich SM, Tandon A, Park D, Chu CT, et al. Loss of PINK1 function promotes mitophagy through effects on oxidative stress and mitochondrial fission. J Biol Chem. 2009; 284: 13843-13855.

30. Bueler H. Impaired mitochondrial dynamics and function in the pathogenesis of Parkinson’s disease. Exp Neurol. 2009; 218: 235-246.

31. McCoy MK, Cookson MR. Mitochondrial quality control and dynamics in Parkinson’s disease. Antioxid Redox Signal. 2012; 16: 869-882.

32. Ebato C, Uchida T, Arakawa M, Komatsu M, Ueno T, Komiya K, et al. Autophagy is important in islet homeostasis and compensatory increase of beta cell mass in response to high-fat diet. Cell Metab. 2008; 8: 325-332.

33. Jung HS, Chung KW, Won Kim J, Kim J, Komatsu M, Tanaka K, et al. Loss of autophagy diminishes pancreatic beta cell mass and function with resultant hyperglycemia. Cell Metab. 2008; 8: 318-324.

34. Las G, Serada SB, Wikstrom JD, Twig G, Shirihai OS. Fatty acids suppress autophagic turnover in β-cells. J Biol Chem. 2011; 286: 42534-42544.

35. Thomas KJ, McCoy MK, Blackinton J, Beilina A, van der Brug M, Sandebring A, et al. DJ-1 acts in parallel to the PINK1/parkin pathway to control mitochondrial function and autophagy. Hum Mol Genet. 2011; 20: 40-50.

36. Inberg A, Linial M. Protection of pancreatic beta-cells from various stress conditions is mediated by DJ-1. J Biol Chem. 2010; 285: 25686- 25698.

37. Geng X, Lou H, Wang J, Li L, Swanson AL, Sun M, et al. α-synuclein binds the K(ATP) channel at insulin-secretory granules and inhibits insulin secretion. Am J Physiol Endocrinol Metab. 2011; 300: E276-286

38. Devi L, Raghavendran V, Prabhu BM, Avadhani NG, Anandatheerthavarada HK. Mitochondrial import and accumulation of alpha-synuclein impair complex I in human dopaminergic neuronal cultures and Parkinson disease brain. J Biol Chem. 2008; 283: 9089- 9100.

39. Winslow AR, Chen CW, Corrochano S, Acevedo-Arozena A, Gordon DE, Peden AA, et al. α-synuclein impairs macroautophagy: implications for Parkinson’s disease. J Cell Biol. 2010; 190: 1023-1037.

40. Steneberg P, Bernardo L, Edfalk S, Lundberg L, Backlund F, Ostenson CG, et al. The type 2 diabetes-associated gene ide is required for insulin secretion and suppression of α-synuclein levels in β-cells. Diabetes. 2013; 62: 2004-2014.

41. Tofaris GK. Lysosome-dependent pathways as a unifying theme in Parkinson’s disease. Mov Disord. 2012; 27: 1364-1369.

42. Arduíno DM, Esteves AR, Oliveira CR, Cardoso SM. Mitochondrial metabolism modulation: a new therapeutic approach for Parkinson’s disease. CNS Neurol Disord Drug Targets. 2010; 9: 105-119.

43. Chien WL, Lee TR, Hung SY, Kang KH, Wu RM, Lee MJ, et al. Increase of oxidative stress by a novel PINK1 mutation, P209A. Free Radic Biol Med. 2013; 58: 160-169.

Shridas P, Zhi L, Akundi RS, Webb NR, Pearson KJ, et al. (2013) PTEN-Induced Kinase 1 Regulates Mitochondrial Integrity and Insulin Secretion in Mouse Pancreatic β –Cells. J Endocrinol Diabetes Obes 1(1): 1007.

Received : 24 Jul 2013
Accepted : 20 Aug 2013
Published : 25 Aug 2013
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X