© 2014. Published by The Company of Biologists Ltd.

Kukic  et  al.,  Page 1 of 33  

1   2   3   4   5   6   7  

Zinc efflux through lysosomal exocytosis prevents zinc-induced toxicity.

8  

Journal of Cell Science

Accepted manuscript

9  

Ira Kukic1, Shannon L. Kelleher2,3,4 and Kirill Kiselyov1

10   11   12  

1

13  

15260 USA,

14  

Development, The Pennsylvania State University, University Park, Pennsylvania 16802, USA,

15  

Department of Surgery, Penn State Hershey Medical Center, Hershey, Pennsylvania 17033,

16  

USA, and

17  

Center, Hershey, Pennsylvania 17033, USA.

From the Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania

4

2

the Department if Nutritional Sciences, College of Health and Human 3

Department of Cellular and Molecular Physiology, Penn State Hershey Medical

18   19   20  

Running title: Lysosomal exocytosis and Zn2+ toxicity  

21   22  

To whom correspondence should be addressed: Kirill Kiselyov, Department of Biological

23  

Sciences, University of Pittsburgh, 519 Langley Hall, 4249 Fifth Avenue, Pittsburgh, PA, 15260.

24  

Tel: 412-624-4317; Fax: 412-624-4759; E-mail: [email protected].

25   26  

Key words: Zinc, Golgi, lysosomes, metallothionein, exocytosis, Zn2+ transport, ZnT, Slc30a.

27   28  

Word count (excluding references): 6421

29  

Figures: 9

30  

Supplementary Figures: 3

31   32  

JCS Advance Online Article. Posted on 14 May 2014

Journal of Cell Science

Accepted manuscript

Kukic  et  al.,  Page 2 of 33  

33  

Summary

34  

Zinc (Zn2+) is an essential micronutrient and an important ionic signal, whose excess as well as

35  

scarcity are detrimental to cells. Free cytoplasmic Zn2+ is controlled by a network of Zn2+

36  

transporters and chelating proteins. Recently, lysosomes became the focus of studies in Zn2+

37  

transport, as they were shown to play a role in zinc-induced toxicity by serving as Zn2+ sinks that

38  

absorb Zn2+ from the cytoplasm. Here we investigate the impact of the lysosomal Zn2+ sink on

39  

the net cellular Zn2+ distribution and its role in cell death. We found that lysosomes play a

40  

cytoprotective role during exposure to extracellular Zn2+. Such a role required lysosomal

41  

acidification and exocytosis. Specifically, we found that the inhibition of lysosomal acidification

42  

using Bafilomycin A1 (Baf) lead to a redistribution of Zn2+ pools, and increased apoptosis.

43  

Additionally, the inhibition of lysosomal exocytosis through knockdown (KD) of the lysosomal

44  

SNARE proteins VAMP7 and Synaptotagmin VII (SYT7) suppressed Zn2+ secretion and

45  

VAMP7 KD cells had increased apoptosis. These data show that lysosomes play a central role in

46  

Zn2+ handling, suggesting a novel Zn2+ detoxification pathway.

47   48  

Introduction

49  

Cellular Zn2+ dyshomeostasis has been linked to a number of human pathologies including

50  

growth defects (Prasad, 2013), impaired immune function (Rink and Gabriel, 2000), diabetes

51  

(Jansen et al., 2009), and neurodegenerative diseases (Forsleff et al., 1999; Rulon et al., 2000;

52  

Lee et al., 2002; Vinceti et al., 2002). Regulation of cellular Zn2+ levels involves controlling its

53  

influx, export and chelation. In general, Zn2+ transport is regulated by ZnT and ZIP transporters,

54  

and it is chelated by Zn2+ binding metallothioneins (MTs).

55   56  

In addition to Zn2+ evacuation across the plasma membrane (PM) by the Zn2+ transporter ZnT1

57  

(Palmiter and Findley, 1995), Zn2+ is exported from the cytoplasm into the organelles by the

58  

dedicated ZnT transporters such as ZnT6 for the Golgi (Huang et al., 2002), and ZnT2 and ZnT4

59  

for the lysosome (Palmiter et al., 1996; Huang and Gitschier, 1997; Falcon-Perez and

60  

Dell'Angelica, 2007; McCormick and Kelleher, 2012). This organellar Zn2+ export lowers

61  

potentially toxic cytoplasmic Zn2+ concentrations in pathophysiological conditions such as

62  

neurodegeneration (Kanninen et al., 2013) and breast cancer (Lopez et al., 2011). Moreover, it

63  

provides Zn2+ to organellar processes that require it, such as the maturation of enzymes like the

Kukic  et  al.,  Page 3 of 33  

64  

lysosomal acid sphingomyelinase (Schissel et al., 1996), and the secretion of Zn2+ under normal

65  

physiological conditions such as synaptic transmission (Frederickson and Bush, 2001) and

66  

lactation (Kelleher et al., 2009)

Journal of Cell Science

Accepted manuscript

67   68  

The upregulation of Zn2+ chelation and transport machinery following the activation of the

69  

transcription factor MTF-1 by Zn2+ binding (Andrews, 2001) requires time for transcription,

70  

translation and protein processing. It is tempting to speculate that Zn2+ export into organelles

71  

serves as a first line of defense to provide temporary Zn2+ storage, giving cells time to upregulate

72  

Zn2+ chelators and transporters. Our recent data on the role of lysosomes in Zn2+ handling, as

73  

well as some recently published results suggest that lysosomes play a role of such Zn2+ sinks,

74  

temporarily storing Zn2+ (Hwang et al., 2008; Kukic et al., 2013). In this paper, we sought to

75  

delineate the role of lysosomes in protection against Zn2+-induced toxicity.

76   77  

Zn2+ is transported from the cytoplasm into lysosomes by ZnT2 and ZnT4 (Palmiter et al., 1996;

78  

Huang and Gitschier, 1997; Falcon-Perez and Dell'Angelica, 2007). Zn2+ can also be delivered to

79  

the lysosomes through endocytosis or autophagy (Lee and Koh, 2010; Cho et al., 2012). What

80  

happens to Zn2+ absorbed by the lysosomes? A recent series of work from several labs indicate

81  

that Zn2+ buildup in the lysosomes is toxic. It leads to lysosomal membrane permeabilization

82  

(LMP), to the release of the lysosomal enzymes such as Cathepsins and to cell death (Hwang et

83  

al., 2008; Chung et al., 2009; Lee et al., 2009; Hwang et al., 2010). As such, the lysosomal Zn2+

84  

accumulation may constitute a cell death mechanism during normal remodeling of Zn2+-rich

85  

tissues, such as the mammary gland (Kelleher et al., 2011), as well as in pathological conditions.

86  

With this in mind, we sought to answer whether or not the accumulation of Zn2+ in the lysosome

87  

is the terminal depot for cellular Zn2+.

88   89  

Alternatively, it is possible that lysosomal Zn2+ dissipates and lysosomes constitute only a

90  

temporary Zn2+ storage site. Our recently published data suggest that the lysosomal ion channel

91  

transient receptor potential mucolipin 1 (TRPML1) is at least partly responsible for dissipating

92  

the lysosomal Zn2+ into the cytoplasm (Kukic et al., 2013). It should be noted that lysosomes fuse

93  

with the PM via a process involving a specific SNARE complex, which includes the VAMP7

94  

protein and SYT7 (Martinez-Arca et al., 2000; Braun et al., 2004; Rao et al., 2004; Logan et al.,

Kukic  et  al.,  Page 4 of 33  

95  

2006; Mollinedo et al., 2006). Such secretion was recently proposed to contribute to excretion of

96  

undigested/indigestible products inside lysosomes (Medina et al., 2011). In the course of the

97  

present studies, we used VAMP7 and SYT7 KD to suppress lysosomal secretion and assess its

98  

role in Zn2+ clearance from the cells.

Journal of Cell Science

Accepted manuscript

99   100  

Here we aimed to establish the functional context of the lysosomal Zn2+ accumulation. Our

101  

findings indicate that lysosomes actively absorb Zn2+ and secrete it across the PM, since

102  

suppressing the lysosomal Zn2+ absorption or secretion causes Zn2+ buildup in the cytoplasm,

103  

Golgi apparatus and mitochondria, leading to apoptosis.

104   105  

Results

106  

Towards testing the role of the lysosomal Zn2+ sink on cellular Zn2+ handling, we blocked the

107  

lysosomal H+ pump in HeLa cells using 1 μM Baf and exposed cells to 100 μM ZnCl2 for 3

108  

hours. The resulting cytoplasmic Zn2+ spikes were measured using live-cell confocal microscopy

109  

and FluoZin-3,AM as described before (Kukic et al., 2013). Figure 1A (Fig 1A) shows that the

110  

exposure of Baf-treated cells to Zn2+ caused a significantly higher FluoZin-3,AM response than

111  

the exposure of untreated cells to Zn2+. Although Baf has been shown to decrease cytoplasmic

112  

pH, potentially affecting Zn2+ binding to cytoplasmic proteins, or FluoZin-3,AM fluorescence,

113  

the magnitude of the observed effects appear to be incompatible with the quantitative estimates

114  

of changes induced by Baf. Thus, Baf’s effect on cytoplasmic pH appears to be small, within

115  

only tenths of pH units (Heming et al., 1995). The degree of pH change necessary to cause an

116  

effect on Zn2+ handling, on the other hand, significantly exceeds that reported to be caused by

117  

Baf. A pH drop below 6.7 is required to trigger an increase in intracellular Zn2+ according to one

118  

set of studies (Kiedrowski, 2012), while another set showed that metallothioneins release Zn2+

119  

only after cytoplasmic pH drops below 5.0 (Jiang et al., 2000). Thus, the increase in cytoplasmic

120  

Zn2+ caused by Baf likely correlates with the loss of lysosomal function, rather than cytosolic pH

121  

changes.

122   123  

We have previously shown that Zn2+ transporters ZnT2 and ZnT4 colocalize with the lysosomal

124  

ion channel TRPML1 in HeLa cells (Kukic et al., 2013). We suggested that these transporters

125  

play a role in loading of the lysosomes with Zn2+. In order to test this assumption, we KD ZnT2

Kukic  et  al.,  Page 5 of 33  

126  

and ZnT4 using siRNA as described before and tested the resulting changes in Zn2+ handling

127  

using FluoZin-3,AM. Fig 1B shows that ZnT2 and ZnT4 KD increased cytoplasmic Zn2+ levels

128  

observed in these cells after 3 hour long treatment with 100 μM ZnCl2. These results are in

129  

agreement with the previously published data on the dependence of ZnTs activity on the acidic

130  

environment of the lysosomes (Chao and Fu, 2004; Ohana et al., 2009) for Zn2+ binding and

131  

transporting activity.

Journal of Cell Science

Accepted manuscript

132   133  

The upregulation of MTF-1–dependent, Zn2+-responsive genes such as MT2a and ZnT1 (Saydam

134  

et al., 2002) indicates elevated cytoplasmic Zn2+. MT2a mRNA was used previously in our

135  

studies of the role of TRPML1 in Zn2+ handling. We measured the expression of the mRNA of

136  

these genes using qRT-PCR (Fig 2). An increase in MT2a and ZnT1 mRNA responses to Zn2+ in

137  

cells treated with Baf is evident. With MT2a mRNA levels in DMSO-treated (no Zn2+) cells

138  

taken for 100%, MT2a mRNA levels were 816.39±73.61% in cells treated with DMSO+Zn2+

139  

(n=4; p

Zn2+ efflux through lysosomal exocytosis prevents Zn2+-induced toxicity.

Zn(2+) is an essential micronutrient and an important ionic signal whose excess, as well as scarcity, is detrimental to cells. Free cytoplasmic Zn(2+)...
3MB Sizes 0 Downloads 0 Views