Iron Oxide Nanoparticles Induce Autophagosome ... - ACS Publications

Jun 10, 2016 - through Multiple Mechanisms: Lysosome Impairment, Mitochondrial. Damage, and ... Fe3O4 nanoparticles extensively impair lysosomes and...
0 downloads 0 Views 2MB Size
Subscriber access provided by UNIV OF NEBRASKA - LINCOLN

Article

Iron oxide nanoparticles induce autophagosome accumulation through multiple mechanism: lysosome impairment, mitochondrial damage and ER stress Xudong Zhang, Hongqiu Zhang, Xin Liang, Jinxie Zhang, Wei Tao, Xianbing Zhu, Danfeng Chang, Xiaowei Zeng, Gan Liu, and Lin Mei Mol. Pharmaceutics, Just Accepted Manuscript • DOI: 10.1021/acs.molpharmaceut.6b00405 • Publication Date (Web): 10 Jun 2016 Downloaded from http://pubs.acs.org on June 13, 2016

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Molecular Pharmaceutics is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 28

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

1

Iron oxide nanoparticles induce autophagosome accumulation

2

through multiple mechanism: lysosome impairment, mitochondrial

3

damage and ER stress

4

5

Xudong Zhang1, 2, 3, a, Hongqiu Zhang1, 2, a, Xin Liang2,4, Jinxie Zhang1, 2,, Wei Tao1,2,

6

Xianbing Zhu1,2 , Danfeng Chang1, 2, Xiaowei Zeng1, 2, Gan Liu1, 2 , & Lin Mei1, 2, *

7 8 9 10 11 12 13

1

School of Life Sciences, Tsinghua University, Beijing 100084, China;

2

Division of Life and Health Sciences, Graduate School at Shenzhen, Tsinghua

University, Shenzhen 518055, P.R. China 3

Joint Department of Biomedical Engineering, University of North Carolina at

Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA 4

Department of Pharmacological and Physiological Science and Center for

Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri, USA

14 15 16 17 18 19

20 21 22

a

These authors contributed equally to this work.

*

Corresponding author.

Tel/Fax: +86 75526036736, E-mail: [email protected]

23 24 25 26 1

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1 2 3

Table of Contents Graphic

4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 2

ACS Paragon Plus Environment

Page 2 of 28

Page 3 of 28

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

1

ABSTRACT

2

Magnetite (iron oxide, Fe3O4) nanoparticles have been widely used for drug

3

delivery and magnetic resonance imaging (MRI). Previous studies have shown that

4

many metal-based nanoparticles including Fe3O4 nanoparticles can induce

5

autophagosome accumulation in treated cells. However, the underlying mechanism is

6

still not clear. To investigate the bio-safety of Fe3O4 and PLGA-coated Fe3O4

7

nanoparticles, some experiments related to the mechanism of autophagy induction by

8

these nanoparticles have been investigated. In this study, the results showed that

9

Fe3O4, PLGA-coated Fe3O4 and PLGA nanoparticles could be taken up by the cells

10

through cellular endocytosis. Fe3O4 nanoparticles extensively impair lysosomes and

11

lead to the accumulation of LC3-positive autophagosomes, while PLGA-coated Fe3O4

12

nanoparticles reduce this destructive effect on lysosomes. Moreover, Fe3O4

13

nanoparticles could also cause mitochondrial damage, ER and Golgi body stresses,

14

which induce autophagy, while PLGA-coated Fe3O4 nanoparticles reduce the

15

destructive effect on these organelles. Thus, the Fe3O4-nanoparticle induced

16

autophagosome accumulation may be caused by multiple mechanisms. The

17

autophagosome accumulation induced by Fe3O4 was also investigated. The Fe3O4,

18

PLGA-coated Fe3O4 and PLGA nanoparticle treated mice were sacrificed to evaluate

19

the toxicity of these nanoparticles on the mice. The data showed that Fe3O4

20

nanoparticle treated mice

21

autophagosomes in the kidney and spleen in comparison to the PLGA-coated Fe3O4

22

and PLGA nanoparticles. Our data clarifies the mechanism by which Fe3O4 induces

23

autophagosome accumulation and the mechanism of its toxicity on cell organelles and

24

mice organs. These findings may have an important impact on the clinical application

25

of Fe3O4 based nanoparticles.

26

Key Words: Fe3O4; PLGA; Autophagy; Lysosome; Nanomedicine

would lead to the extensive accumulation of

27 28 29 30 31 3

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

Page 4 of 28

INTRODUCTION

2

Magnetite (Fe3O4) is a miraculous half-metallic material because it is magnetic.

3

The external magnetic field allows Fe3O4 particles localize to a specific area, which

4

makes Fe3O4 a promising material for medicine [1]. Moreover, nanosized Fe3O4

5

changes some of these magnetic properties, such as making the nanoparticles own the

6

superparamagnetic phenomena [1, 2]. Superparamagnetic nanoparticles have many

7

promising applications in nanomedicine such as cell targeting, drug delivery,

8

magnetic resonance imaging (MRI), hyperthermia, magnetofection and tissue repair

9

[1]. Before the application to medicine, the particles have to be modified with some

10

molecular layers to generate biocompatibility [3]. Inorganic materials such as gold,

11

silica, and alumina have been extensively used to modify the Fe3O4 nanoparticles [1].

12

Some organic polymeric materials based on poly(ethylene-co-vinyl acetate),

13

poly(vinylpyrrolidone)

14

poly(ethyleneglycol) (PEG), and poly(vinyl alcohol) (PVA) are also used to modify

15

the Fe3O4 nanoparticles [1]. Linking ligands such as antibodies, proteins, peptides and

16

small molecules to the polymer surfaces will make the particles target specific tissues

17

and organs for drug delivery and diagnosis [4].

(PVP),

poly(lactic-co-glycolic

acid)

(PLGA),

18

Autophagy involves the degradation of deleterious cellular components including

19

aggregation-prone proteins and damaged organelles or invaded pathogens [5, 6]. An

20

isolated double membrane (also termed phagophore), sequesters the cytoplasm

21

components, including several organelles and cytosol, to form a vesicle (also referred

22

as autophagosome) [6]. The autophagosome is fused with a lysosome and forms an

23

auto-lysosome which degrades the “cargo” within it. Many previous publications have

24

reported that autophagosomes accumulate within the cells after treating the cells with

25

metal-based NPs such as gold NPs [7, 8], ferric oxide (Fe2O3 and Fe3O4) [9],

26

manganese oxide (MnO), copper oxide (CuO) [10], neodymium oxide (Nd2O3) [11],

27

titanium oxide (TiO2) [12] and silica oxide (SiO2) [10, 12]. Many cellular stressed

28

activities could induce the initiation of autophagy in the cells. These stressful events

29

include ER stress, Golgi body stress and mitochondrial damage [13]. Most of these 4

ACS Paragon Plus Environment

Page 5 of 28

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

1

NPs are found to be internalized by cells through endocytosis [4]. Internalization

2

allows the NPs reach the subcellular locations and interact with diverse organelles

3

such as mitochondria, Endoplasmic Reticulum (ER), lysosome, Golgi body, nuclear

4

membrane, even chromosomes [4]. Thus, many cellular reactions including autophagy

5

may be induced by interactions between the NPs and organelles. Thus, the presence of

6

autophagosomes with the cell-induced metal-based nanoparticles may be caused by

7

multiple mechanisms.

8

However, Autophagy induction cannot only determined by the presence of

9

autophagosome within the cells. Autophagy is a complex cellular process, the

10

autophagosome is matured from the isolated membrane, and it finally fuse with

11

lysosome,

12

autophagosomes can result from true induction of autophagy or blockade of the

13

turnover of autophagosome by lysosomes [14]. Previous research shows that

14

autophagosomes are induced by treating the cells with the gold NPs [7]. However, a

15

recent report shows that gold NPs could impair the function of the lysosomes, and

16

lead to the accumulation of autophagosomes [15]. In other words, the gold NPs may

17

not induce autophagy, instead they may impair the autophagic flux [15]. Autophagic

18

substrates such as p62/SQSTM1 are required for establishing induction of autophagy

19

[5]. When the protein levels are decreased, it indicates that autophagy may be induced.

20

Conversely, when the protein level of p62 is increased, the autophagic flux may be

21

blocked and the function of the lysosome may be impaired. Our work investigated the

22

detailed relationship and mechanism between Fe3O4 nanoparticles and autophagy,

23

which may have important implications for the development of Fe3O4 nanoparticles

24

for biomedical applications.

25

EXPERIMENTAL SECTION

26

Materials

which

called

“autophagic

flux’’ [14].

Thus,

accumulation

of

27

Coumarin-6, oleic acid, PLGA (acid terminated, lactide:glycolide 50:50,

28

Mw24,000-38,000), oleylamine and Fe(acac)3 were purchased from Sigma-Aldrich

29

(St. Louis, MO, USA). All other chemicals of highest quality were commercially

30

available and used as received. Antibodies against EEA1, LC3, p62, mTOR, p-mTOR, 5

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

ULK1 and p-ULK1 were from Cell Signaling Technology (Beverly, MA, USA).

2

Antibodies against Tom20 were from Santa Cruz Biotechnologies. Antibody against

3

actin was obtained from Abmart, Inc. (Shanghai, China). Golgi’s Complex Tracker

4

Red, ER’s Tracker Red and Lysosome Tracker Red probes were obtained from

5

Beyotimes (Jiangsu, China). JC-1 and ROS probes were also obtained from

6

Beyotimes. The EGFP-LC3 and DsRed-LC3 plasmids were from our lab.

7

DsRed-Rab5 and DsRed-Rab7 were from Addgene (Cambridge, MA, USA). All

8

plasmids were confirmed by automated DNA sequencing. Cells were transiently

9

transfected with the plasmids using lipofectamine 2000 (Invitrogen) according to the

10

manufacturer’s instructions.

11

Methods

12

Synthesis of Fe3O4

13

The iron oxide (Fe3O4) nanoparticles were prepared by solvent-free thermal

14

decomposition method of Fe(acac)3 as done earlier. 2 mM of Fe(acac)3 was solved in

15

a mixture of 10 ml oleic acid (90%, sigma-aldrich) and 10 ml oleylamine (70%,

16

sigma-aldrich). Then the solution is magnetically stirred under a flow of argon gas. It

17

was heated at 120 °C for 1 hour for dehydration, then heated to 300 °C quickly and

18

kept it for 1 hour. The solution was cooled in room temperature for hours until it is

19

down to room temperature. Then 20 ml of ethanol was added and IOs were collected

20

by centrifugation at 10,000 rpm. After that, it is washed by ethanol three times.

21

Finally the Fe3O4 particles were dispersed in THF.

22

Formulation of Fe3O4-encapsulated and coumarin-6-loaded PLGA nanoparticles

23

The IOs (Fe3O4)-encapsulated PLGA capsules were prepared using the

24

membrane dialysis method [29]. Briefly, 100 mg of copolymer PLGA and 10 mg of

25

IOs were dissolved in 10 mL of THF. With this mixture, 15 mL of deionized water

26

was added dropwise under stirring. The resulting solution was transferred to dialysis

27

bag (MWCO: 3500 Da, Spectra/Por® 6, Spectrum Laboratories, CA, USA) and

28

dialyzed against deionized water for 24 h. Finally, these capsules were freeze-dried

29

for two days. In addition, the coumarin-6-loaded PLGA capsules were prepared in the

30

same way except the Fe3O4 was replaced by coumarin-6. 6

ACS Paragon Plus Environment

Page 6 of 28

Page 7 of 28

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

1

Characterization of Fe3O4, PLGA-coated Fe3O4 and PLGA NPs

2

The size and zeta potential of the nanoparticles were measured by Malvern

3

Mastersizer 2000 (Zetasizer Nano ZS90, Malvern Instruments Ltd., UK). The

4

morphology of the nanoparticles was observed by transmission electron microscopy

5

(TEM, Tecnai G2 20, FEI Company, Hillsboro, Oregon, USA). Before observation,

6

the sample was deposited onto a copper grid coated with carbon and dried at room

7

temperature.

8

Cell culture

9

MCF-7 cells were cultured in Dulbecco’s Modified Eagle’s Medium (DMEM)

10

supplemented with 10% Fetal Bovine Serum (FBS).

11

Cellular uptake of coumarin-6-loaded PLGA-coated Fe3O4 and PLGA NPs.

12

Coumarin-6 was used as a model fluorescent molecule, which was formulated in

13

PLGA-coated Fe3O4 and PLGA NPs. Cellular uptake of the coumarin-6 loaded

14

PLGA-coated Fe3O4 NPs and PLGA NPs by MCF-7 cells were assessed. The

15

non-transfected or DsRed-Rab5 and DsRed-Rab7 cells were incubated with 100

16

µg/mL coumarin-6-loaded PLGA-coated Fe3O4 and PLGA NPs at 37 ℃ for 2 h. For

17

lysosome detection, the cells were incubated with Lyso-Tracker Red for 1 h. Then the

18

cells were washed with PBS for three times, and then fixed by 4% paraformaldehyde

19

for 20 min. Cells were stained with DAPI for 15 min and washed three times with

20

PBS. Confocal microscopy was performed on a FLUO-VIEW laser scanning confocal

21

microscope (Olympus, FV1000, Olympus Optical, and Japan) in sequential scanning

22

mode using a 60~100×objective.

23

Autophagy assays

24

The cells were transfected with EGFP-LC3 under indicated conditions and then

25

fixed in 4% paraformaldehyde. The percentage of cells with fluorescent dots

26

representing EGFP-LC3 translocations were counted by confocal microscopy as

27

described previously [31].

28 29 30

LC3II protein level was detected using the anti-LC3 antibody. Immunoblotting Immunoblotting analysis was performed as previously described [31]. For 7

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

abbreviation, cell lysates were resolved on 12% SDS-PAGE and analyzed by

2

immunoblotting using LC3, p62, mTOR, p-mTOR, ULK1 and p-ULK1 antibody,

3

followed by enhanced chemiluminescence (ECL) detection (Thermo Scientific).

4

Mitochondrial transmembrane potential (∆Ψm) detection

5

JC-1 mitochondrial membrane potential assay kit was used to detect the

6

mitochondrial (∆Ψm) according to the manufacturer’s protocol. Briefly, the MCF-7

7

cells were seeded in 12-well plates, and treated with 100 µg/mL Fe3O4 NPs,

8

PLGA-coated Fe3O4 (PLGA-Fe3O4) NPs and PLGA NPs for 24 h. Then, 100 µl JC-1

9

staining solution was add to each well and incubated for another 15~30 min. The cells

10

were washed with PBS for three times, and then fixed by 4% paraformaldehyde for 20

11

min. After that, cells were stained with DAPI for 15 min and washed three times with

12

PBS. Confocal microscopy was performed on a FLUO-VIEW laser scanning confocal

13

microscope (Olympus, FV1000, Olympus Optical, and Japan) in sequential scanning

14

mode using a 60~100×objective.

15

Reactive oxygen species (ROS) detection

16

MCF-7 cells were seeded in 6-well plates. The cells were treated with 100 µg/mL

17

Fe3O4 NPs, PLGA-coated Fe3O4 (PLGA-Fe3O4) NPs and PLGA NPs for 24 h, Then

18

cells were incubated with DCFH-DA (Beyotime Biotechnology, China) according to

19

the instruction. For quantitative analysis, the ROS levels were examined using a

20

Partec PAS III flow cytometer (Partec, Munster, Germany).

21

In vivo autophagy assay with NIH mice

22

Animal experiments were approved by the Administrative Committee on Animal

23

Research in the Tsinghua University Shenzhen Graduate School. Female severe

24

combined NIH mice were purchased from Guangdong Medical Laboratory Animal

25

Center. Mice were detected by organ-size and weight. IOs (at a single dose of 10 mg

26

IOs /kg), PLGA-coated Fe3O4 NPs (at a single dose of 10 mg IOs /kg) and PLGA NPs

27

were injected via peritoneal cavity in PBS on days 0, 2, 4, 8, 10 and 12. Mice were

28

sacrificed by cervical decapitation 20 days after treatment. The terminal organ

29

damage was measured and utilized to evaluate the autophagy. The organ samples were

30

used to make LC3. 8

ACS Paragon Plus Environment

Page 8 of 28

Page 9 of 28

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

1

Immunofluorescence assay

2

Immunohistochemical analysis was performed as previously described [20]. For

3

abbreviation, the cells or tumor sections were incubated with primary antibodies. The

4

TRITC and FITC labeled secondary antibody was used to detect the primary antibody.

5

Statistical methodology

6

All results are reported as the mean ± SD of three independent experiments.

7

Comparisons were performed using a two-tailed paired Student’s t test (* P