Inhibiting microRNA-144 abates oxidative stress and reduces apoptosis in hearts of streptozotocin-induced diabetic mice Manli Yu, Yu Liu, Bili Zhang, Yicheng Shi, Ling Cui, Xianxian Zhao PII: DOI: Reference:

S1054-8807(15)00077-0 doi: 10.1016/j.carpath.2015.06.003 CVP 6849

To appear in:

Cardiovascular Pathology

Received date: Revised date: Accepted date:

1 April 2015 16 June 2015 17 June 2015

Please cite this article as: Yu Manli, Liu Yu, Zhang Bili, Shi Yicheng, Cui Ling, Zhao Xianxian, Inhibiting microRNA-144 abates oxidative stress and reduces apoptosis in hearts of streptozotocin-induced diabetic mice, Cardiovascular Pathology (2015), doi: 10.1016/j.carpath.2015.06.003

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Inhibiting microRNA-144 abates oxidative stress and reduces apoptosis in hearts of

RI P

T

streptozotocin-induced diabetic mice

SC

Manli Yu*, Yu Liu*, Bili Zhang, Yicheng Shi, Ling Cui, Xianxian Zhao

University, Shanghai, China

MA NU

Department of Cardiovasology, Changhai Hospital, Second Military Medical

*The first two authors contributed equally to the work.

ED

Correspondence to Xianxian Zhao, Department of Cardiovasology, Changhai Hospital,

PT

Second Military Medical University, Changhai Road 168, Shanghai, 200433, China Tel: +8602131161245; Fax: +8602131161263;

AC

CE

E-mail: [email protected]

Summary:

Inhibiting microRNA-144 abated oxidative stress, reduced apoptosis, and improved cardiac function in STZ-induced diabetic mice, possibly via enhancing Nrf2 expression.

1

ACCEPTED MANUSCRIPT ABSTRACT: Introduction: Hyperglycemia-induced reactive oxygen species (ROS) generation

RI P

T

contributes to the development of diabetic cardiomyopathy. However, little is known about the role of microRNAs (miRNAs) in the regulation of ROS formation and

SC

myocardial apoptosis in streptozotocin (STZ)-induced diabetic mice. Methods and Results: It was observed that microRNA-144 (miR-144) level was

MA NU

lower in heart tissues of STZ-induced diabetic mice. High glucose exposure also reduced miR-144 levels in cultured cardiomyocytes. Moreover, miR-144 modulated high glucose-induced oxidative stress in cultured cardiomyocytes by directly targeting

ED

nuclear factor-erythroid 2-related factor 2 (Nrf2), which was a central regulator of

PT

cellular response to oxidative stress. The miR-144 mimics aggravated high glucose-induced ROS formation and apoptosis in cardiomyocytes, which could be

CE

attenuated by treatment with Dh404, an activator of Nrf2. Meanwhile, inhibition of

AC

miR-144 suppressed ROS formation and apoptosis induced by high-glucose in cultured cardiomyocytes. What was more important, reduced myocardial oxidative stress and apoptosis and improved cardiac function were identified in STZ-induced diabetic mice when treated with miR-144 antagomir. Conclusion: Although miR-144 cannot explain the increased oxidative stress in STZ, therapeutic interventions directed at decreasing miR-144 may help to decrease oxidative stress in these hearts. Inhibition of miR-144 might have clinical potential to abate oxidative stress as well as to reduce cardiomyocyte apoptosis, and improve cardiac function in diabetic cardiomyopathy.

2

ACCEPTED MANUSCRIPT

Keywords: streptozotocin, diabetic cardiomyopathy, microRNA-144, nuclear

AC

CE

PT

ED

MA NU

SC

RI P

T

factor-erythroid 2-related factor 2, oxidative stress

3

ACCEPTED MANUSCRIPT INTRODUCTION Diabetes mellitus has reached an epidemic level worldwide, with a prevalence of 4%

RI P

T

in 1995 and an anticipated prevalence of 5.4% in 2025, corresponding to 365 million people suffered from diabetes in 2011 and this number is expected to rise up to 552

SC

million by 2030 [1]. Diabetic cardiomyopathy is responsible for higher incidence of

the diabetic population [2].

MA NU

sudden cardiac death and represents the leading cause of morbidity and mortality in

Apoptotic cell death is increased in the diabetic heart of patients and animal models [3, 4]. In diabetic cardiomyopathy, apoptosis as a comprehensive consequence of cardiac

ED

responses to various stresses causes a loss of contractile tissue, which initiates a

PT

cardiac remodeling and fibrosis; therefore, the cardiac apoptosis has been identified as a pivotal cause of various cardiomyopathies [5, 6]. Among apoptotic stimuli, cardiac

CE

ROS formation was closely related to apoptosis [7]. Oxidative stress occurred in

AC

diabetic hearts from both type 1 and type 2 diabetes [8, 9]. Treatment with antioxidants, such as vitamin E, N-acetyl-L-cysteine, and metallothionein, protected cardiomyocytes from apoptosis in high glucose conditions, and prevented diabetic cardiomyopathy and enhanced survival of diabetic animals [10-12]. MicroRNAs (miRNAs, miRs) are an evolutionarily conserved class of small noncoding RNAs of 22-24 nucleotides in length, that act as post-transcriptional regulators of gene expression by binding to the 3’-untranslated region (3’-UTR), finally induce mRNA degradation and/or translational repression in diverse biological processes [13]. The search for regulatory nucleotide sequences that have specific gene

4

ACCEPTED MANUSCRIPT targets have put miRNAs at the forefront of development of therapeutics, and may serve as valuable diagnostic and/or therapeutic targets. Therefore, it is of crucial

RI P

T

importance to gain insight into the role of miRNAs in diabetic cardiomyopathy which will help clarify the molecular mechanisms as well as lead to the development of

SC

novel and effective therapeutic interventions for diabetic cardiomyopathy. The miR-144 was found down-regulated in left ventricles of streptozotocin

MA NU

(STZ)-induced type I diabetic mice by RT-PCR analysis in this work. Using a combination of gain- and loss-of-function studies, we investigated the role of miR-144 on regulation of oxidative stress and apoptosis in cardiomyocytes exposed to

METHODS

CE

Ethical statement

PT

ED

high glucose in vitro and diabetic cardiomyopathy induced by STZ in vivo.

AC

All the animals used in this work received humane care in compliance with institutional animal care guidelines, and were approved by the Local Institutional Committee. All protocols were conducted in accordance with the Guidance Suggestions for the Care and Use of Laboratory Animals, formulated by the Ministry of Science and Technology of China.

Study design Experiment 1: Diabetes was induced in adult male mice by consecutive peritoneal injection of streptozotocin (two group, control and STZ group, n=9 in each group). 8

5

ACCEPTED MANUSCRIPT weeks later, heart tissues were collected for determination of miR-144 expression. Mouse cardiomyocyte cells were incubated in either normal glucose (5 mmol/l) or

RI P

T

high glucose (33 mmol/l). 48 hours later, cells were collected for determination of miR-144 expression.

SC

Experiment 2: Mouse cardiomyocyte cells were transfected with 40 nM miRNA mimic (miR-144) or with 60 nM miRNA inhibitor (anti-miR-144) (Dharmacon)

MA NU

utilizing RNAimax (Invitrogen). 48 hours later, the cells were then incubated in either normal glucose (5 mmol/l) or high glucose (33 mmol/l). 48 hours later, the Nrf2 protein expression was determined by Western blot analysis.

ED

Experiment 3: Mouse cardiomyocyte cells were transfected with 40 nM miRNA

PT

mimic (miR-144) utilizing RNAimax (Invitrogen). 48 hours later, the cells were then incubated in either normal glucose (5 mmol/l) or high glucose (33 mmol/l), and

CE

treated with Dh404 (an Nrf2 activator, 0.2μM). 48 hours later, intracellular ROS and

AC

apoptosis were assayed by DCFH-DA and TUNEL, respectively. Experiment 4: Mouse cardiomyocyte cells were transfected with 60 nM miRNA inhibitor (anti-miR-144). 48 hours later, the cells were then incubated in either normal glucose (5 mmol/l) or high glucose (33 mmol/l). 48 hours later, intracellular ROS and apoptosis were assayed by DCFH-DA and TUNEL, respectively. Experiment 5: Diabetes was induced in adult male mice by consecutive peritoneal injection of streptozotocin. 7 days later, mice were randomized into four groups (n=30-34 in each group). Control and miR-144 antagomir (anti-miR-144) and injected via the tail vein with either a scrambled anti-miR-144 or anti-miR-144 at a dose of 20

6

ACCEPTED MANUSCRIPT mg/kg in 0.2 ml saline twice a week. 7 weeks later, cardiac function was measured

RI P

T

and left ventricles were removed for determination of oxidative stress and apoptosis.

Animal model

SC

C57BL/6 mice (male, 2 months old) were purchased from the Sino-British SIPPR/BK Lab Animal Ltd (Shanghai, China) and housed two per cage under controlled

MA NU

temperature (23-25°C), humidity (50%) and lighting (12-hour light/dark cycle) with food and water provided ad libitum.

Mice were given streptozotocin (STZ) (150 mg/kg i.p.; Sigma-Aldrich, St. Louis, MO,

ED

USA) to induce diabetes, whereas control mice were injected with the same amount of

PT

citrate buffer. 3 days after the injection of STZ, blood was obtained from the tail-vein and glucose levels were measured using the OneTouch Ultra 2 blood glucose

CE

monitoring system (LifeScan, Milpitas, CA, USA). Mice were considered diabetic

AC

and used for the study only if they had hyperglycemia (≥16.7 mM). The control and diabetic mice both raised on standard food and water for the whole experimental period.

Cardiomyocyte isolation and culture Cardiomyocytes were cultured as described previously [14]. Briefly, hearts from adult C57BL/6 mice were excised under deep ether anesthesia and mounted on the cannula of a Langendorff perfusion system. Cardiomyocytes were isolated via perfusion with collagenase, followed by mincing, filtering and transfer to culture medium M199

7

ACCEPTED MANUSCRIPT supplemented with carnitine (2 mM), creatine (5 mM) and taurine (5 mM). Normal glucose: 5 mmol/l, the osmolality were equal by adding 28 mmol/l of mannitol; high

RI P

T

glucose: 33 mmol/l.

SC

Real-time RT-PCR quantification of microRNA expression

Total RNA was isolated from heart tissues or cultured cells using the miRNeasy Mini

MA NU

kit (Qiagen), according to the manufacturer’s protocol. The concentration of RNA was determined by a NanoDrop ND-1000 Spectrophotometer (NanoDrop Tech., Rockland, DE). To detect and quantify mature miR-144 expression, we used TaqMan miRNA

ED

assays (Applied Biosystems) according to manufacturer’s directions and as previously

PT

described [15]. Briefly, 500ng total RNA was used in each reaction and mixed with the RT primer (total 15 μl). The reverse transcription reaction was carried out and 1.33

CE

ul of cDNA was used for PCR reaction along with TaqMan primers (20ul total).

AC

Real-time PCR reactions, including no-template controls, were conducted in triplicate using the ABI 7500 real-time PCR system. Results were analyzed and expressed as CT (threshold cycle) values, and relative expression was calculated using the comparative CT method [16]. Unless otherwise specified, we compared relative levels of microRNA with probes specific for the indicated mature microRNA using total RNA and normalized by evaluating U6 expression.

Luciferase reporter assay The 3’untranslated region (UTR) of the Nrf2 was amplified using primers (forward:

8

ACCEPTED MANUSCRIPT 5’-atttaggaggatttgacc-3’; reverse: 5’-tttttgccagagctaaacaattt-3’) and cloned into the XbaI site downstream of the firefly luciferase gene in the pGL3-control vector

RI P

T

(Promega). The Nrf2 3’UTR mutant reporters were constructed with QuikChange II Site-Directed Mutagenesis (Stratagene). Expression constructs encoding miR-144

SC

were created by insertion into a cytomegalovirus-based pcDNA3 cloning vector (Invitrogen).

MA NU

For Nrf2-3′UTR assay, cardiomyocytes were cotransfected in 12-well plates using the DharmaFECT Duo Transfection Reagent according to the protocol of the manufacturer, with 0.4 μg of the Nrf2-3′UTR luciferase reporter vector and 0.08 μg of

ED

the control vector pMIR-REPORT (Ambion, Texas, USA). For each well, 100nM

PT

miR-144 or scrambled miR control was used. Cell lysates were prepared 72 hous later, and luciferase activity was measured, and expressed as relative light units using a

CE

luciferase assay kit (Promega, Madison, WI, USA). β-galactosidase activity was

AC

measured with a commercially available kit (Promega). 3′UTR activity of each construct was expressed as the ratio of luciferase/β-galactosidase activity. All transfections were performed in triplicate from three independent experiments.

ROS assay in vitro Cells were seeded in a 6-well plate. After treatment for 48 hours, cells were gently washed twice with PBS. Intracellular ROS were quantified by employing ROS-sensitive dye, DCFH-DA (10μM, Beyotime, Shanghai, China). Fluorescent images were acquired from a laser confocal microscope (Zeiss LSM 510 META), and

9

ACCEPTED MANUSCRIPT the intensity on regions of interest was measured.

RI P

T

Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) Assay in vitro

SC

The in situ TUNEL cell death detection kit was used to detect apoptotic cells according to manufacturer’s instructions (Beyotime, Shanghai, China). Briefly, cells

MA NU

were fixed with 4% paraformaldehyde and permeabilized by 0.3% Triton X-100, and then washed twice with PBS. DNA breaks were labeled by incubation (1 hour, 37°C) with terminal deoxynucleotidyltransferase and nucleotide mixture containing

ED

fluorescein isothiocyanate-conjugated dUTP. Cells were nuclear stained with

PT

4’,6-diamidino-2-phenylindole fluorescent dye (DAPI), and the TUNEL positive and

AC

Japan).

CE

total nuclei were observed under a laser scanning confocal microscope (Nikon,

Western blotting

The cells were gently washed twice with iced PBS and harvested at 4℃ in lysis buffer containing 50 mM Tris (pH 7.5), 300 mM NaCl, 1% Triton X-100, 2 mM EDTA, 1 mM PMSF and 2 μM leupeptin. The left ventricles were homogenized in ice cold buffer (0.15 M NaCl, 5 mM EDTA, 10 mM Tris-Cl, 1% Triton X-100 and protease inhibitors cocktail). The homogenates were centrifuged (12,000× g for 15 min at 4°C) and the supernatants were collected. The protein concentration was determined with bovine serum albumin as a standard

10

ACCEPTED MANUSCRIPT by a Bradford assay. Equal amount of protein preparations were run on SDS-polyacrylamide gels, electrotransferred to polyvinylidine difluoride membranes,

RI P

T

and blotted with a primary antibody against Nrf2 (Abcam, San Francisco, CA, USA), cleaved caspase-3 (Abcam, San Francisco, CA, USA), and Pro-caspase-3 (Abcam,

SC

San Francisco, CA, USA) to detect their protein levels, with HRP-conjugated monoclonal antibody against GAPDH (Sigma) serving as a control. Immunoreactive

MA NU

bands were detected by a chemiluminescent reaction (ECL kit, Amersham Pharmacia, USA).

ED

Measurement of cardiac total ROS formation

PT

Total ROS production was detected as the method described by Elks [17]. Briefly, the electron paramagnetic resonance (EPR) measurement was performed with a

CE

BenchTop EPR spectrophotometer e-scan R (Noxygen Science Transfer and

AC

Diagnostics, Elzach, Germany). The spin probe, 1-Hydroxy-3-methoxycarbonyl2,2,5,5-tetramethyl-pyrrolidine (CMH), was used for EPR studies. Small portions (15-20 mg each) of left ventricles from each mice were minced and placed into four wells of a 24-well tissue culture plate containing 20μM Krebs-HEPES buffer with defferoxamine and diethyldithiocarbamate (metal chelators). Tissue pieces were then washed twice with the same buffer to remove any trace contamination. Samples were incubated at 37℃ with 6.6 μl of CMH (200 mM, Enzo Life Sciences, San Diego, USA) for 30 min for ROS measurement.

11

ACCEPTED MANUSCRIPT Measurement of malondialdehyde (MDA) MDA concentration is a presumptive marker of oxidant-mediated lipid peroxidation.

RI P

T

Ventricular homogenates were used for the determination of MDA using a kit

SC

(Cayman, Ann Arbor, USA).

TUNEL assay in vivo

MA NU

Left ventricles were fixed in 4% formaldehyde and paraffin embedded, and 5-μm sections were prepared. TUNEL analysis was performed with a commercially available kit (Dead End Colorimetric TUNEL System) according to the

ED

manufacturer's instructions (ROCHE, Mannheim, DE, USA). The slides were

PT

counterstained with hematoxylin. Three midventricular sections (from the apex to the base) of each heart tissue were analyzed. Cardiomyocyte nuclei were quantified by

CE

randomly counting 10 fields/section. The apoptotic index (percentage of apoptotic

AC

nuclei) was calculated as apoptotic nuclei/total nuclei counted ×100. The evaluation was conducted by an investigator blinded to the study groups.

Cardiac function measurement Transthoracic echocardiography was performed noninvasively with a Vevo 770 high-resolution imaging system equipped with a 30-MHz transducer (RMV-707B; VisualSonics, Toronto, Canada). Mice were lightly anesthetized (0.3 mL of a cocktail containing 100 mg/ml ketamine and 10 mg/ml acepromazine given i.p.) for the duration of the recordings. The heart rate was monitored simultaneously by

12

ACCEPTED MANUSCRIPT electrocardiography (ECG). Left ventricular (LV) end diastolic diameter (LVEDD) and end systolic diameter (LVESD) were used to calculate fractional shortening by

RI P

T

the following formula: Fractional shortening (%) = [(LVEDD – LVESD)/LVEDD] ×100%. LV end diastolic volume (LVEDV) and end systolic volume (LVESV) were

SC

calculated as described previously [18]. Ejection fraction was calculated by the following formula: Ejection fraction (%) = [(LVEDV – LVESV)/LVEDV] ×100%. All

MA NU

echocardiographically derived measures were obtained by averaging the readings of three consecutive beats.

Hearts were isolated and perfused on a Langendorff-system. Maximal and minimal

ED

first derivatives of force (+dF/dtmax and –dF/dtmin) as the rate of contraction and

PT

relaxation were analyzed by PowerLab Chart program (ADInstruments) as described

CE

in previous study [19, 20].

AC

Statistical analysis

All the data are presented as mean ± standard deviations. Comparison between two groups was analyzed using paired Student’s t-test. Comparison among groups was analyzed using a two-way analysis of variance followed by Bonferroni t-test. P < 0.05 was considered statistically significant. Statistical analysis was performed using SPSS 11.0.0 software (SPSS Inc., Chicago, IL, USA).

RESULTS Expression of miR-144 in hearts of STZ-induced diabetic mice

13

ACCEPTED MANUSCRIPT Quantitative RT-PCR results demonstrated significantly lower levels of miR-144 in

RI P

T

hearts of STZ-induced diabetic mice (Fig. 1), when compared to control mice.

Nrf2 is a direct target of miR-144

SC

To understand the potential functional connection between miR-144 and oxidative stress in diabetic cardiomyopathy, we analyzed its predicted target gene. Recent

MA NU

studies have documented that Nrf2 was repressed by miR-144 in K562 cells [21] and cerebromicrovascular endothelial cells [22]. To validate whether miR-144 directly recognized the 3’-UTR of Nrf2, we generated serial constructs harboring the 3’UTR

ED

segment of Nrf2 and its mutant fused downstream to the luciferase coding sequence.

PT

Co-transfection with miR-144 strongly inhibited the luciferase activity; whereas no effect was observed in cells co-transfected with a construct containing a mutated

CE

segment of Nrf2 3’UTR (Fig. 2A).

AC

High glucose reduced miR-144 levels in cultured cardiomyocytes (Fig. 2B). High glucose down-regulated the expression of Nrf2 in cultured cardiomyocytes. More strikingly, protein levels of Nrf2 was further reduced when transfected with miR-144 mimic (Fig. 2C) and enhanced when transfected with miR-144 inhibitor (Fig. 2D). When exposed to normal glucose, transfection with miR-144 mimic or inhibitor had no significant effect on Nrf2 protein expression.

High glucose-induced oxidative stress and apoptosis Transfection with miR-144 mimic aggravated high glucose-induced ROS formation

14

ACCEPTED MANUSCRIPT (Fig. 3A) and apoptosis (Fig. 3B, C) in cultured cardiomyocytes. Treatment with Nrf2 activator-Dh404 fully reversed the effect of miR-144 mimic. When exposed to normal

RI P

T

glucose, neither miR-144 mimic nor Dh404 affected ROS formation or apoptosis. Transfection with miR-144 inhibitor attenuated ROS formation (Fig. 3D) and

SC

apoptosis (Fig. 3E) induced by high glucose in cultured cardiomyocytes.

MA NU

Oxidative stress

In STZ-induced diabetic mice, Nrf2 protein expression (Fig. 4A) was down-regulated, ROS formation (Fig. 4B) and MDA production (Fig. 4C) was enhanced in left

ED

ventricles.

PT

According to the results obtained from in vitro studies, miR-144 antagomir was employed for treatment of STZ-induced cardiomyopathy. Treatment with miR-144

CE

antagomir up-regulated Nrf2 protein expression, suppressed ROS formation, and

AC

reduced MDA content.

Apoptosis

In STZ-induced diabetic mice, caspase-3 (Fig. 5A) was activated in left ventricles when compared to control mice. TUNEL results (Fig. 5B) further validated that apoptosis was enhanced in left ventricles of STZ-induced diabetic mice than that in control mice. Treatment with miR-144 antagomir reduced caspase-3 activation and TUNEL positive cells in left ventricles of STZ-induced diabetic mice.

15

ACCEPTED MANUSCRIPT Cardiac function As shown in Table 1, treatment with miR-144 antagomir had no significant effect on

RI P

T

body weight or levels of fasting blood glucose. When compared to control mice, STZ-diabetic mice showed a significant reduction of +dF/dtmax, -dF/dtmin, fractional

SC

shortening, and ejection fraction. Importantly, myocardial function of diabetic mice

MA NU

was significantly improved when treated with miR-144 antagomir.

DISCUSSION

Diabetic hyperglycemia promotes the production of ROS to lead to oxidative stress

ED

and apoptosis responsible for progressive deterioration of the structure and function of

PT

organs [23, 24]. These findings indicated that factors and pathways regulating ROS production and the cellular redox state play a key role in the progression of diabetes

CE

and diabetes complications including cardiomyopathy [25]. Cells possess an array of

AC

antioxidant defense machinery to prevent or counterbalance damage caused by reactive radicals. One central regulator of antioxidant response is Nrf2, a basic leucine zipper transcription factor. Under oxidative stress, Nrf2 binds to the antioxidant response element (ARE) [26, 27], which is important for the coordinately inducible expression of antioxidant enzymes such as superoxide dismutase, catalase, GPX1, phase II detoxification enzymes such as NAD(P)H: quinone oxidoreductase (NQO1), and glutathione synthesis [28]. Nrf2 is critical in defense against oxidative damage induced by high glucose in cardiomyocytes [29]. STZ-induced diabetes developed more rapidly and severely in Nrf2 KO than WT mice and diabetes induced oxidative

16

ACCEPTED MANUSCRIPT stress and apoptosis in hearts were aggravated in Nrf2 KO mice [30], which indicated the key role of Nrf2 in STZ-induced diabetic cardiomyopathy. It was found that Nrf2

observed in hearts of STZ-induced mice in this work.

RI P

T

expression was down-regulated in human diabetic hearts [31], which was also

SC

Recent studies have documented that Nrf2 was repressed by miR-144 in K562 cells [21] and cerebromicrovascular endothelial cells [22]. In this work, it was validated

MA NU

that Nrf2 was a direct target of miR-144 in adult cardiomyocytes, and miR-144 inhibitor enhanced Nrf2 expression in cells exposed to high glucose in vitro and diabetic hearts in vivo. In addition, miR-144 mimic aggravated high glucose-induced

ED

oxidative stress and apoptosis, and the effect was reversed by treatment with Nrf2

PT

activator, indicating the key role of Nrf2 in the effect of miR-144. Importantly, treatment with miR-144 inhibitor abated oxidative stress, attenuated

CE

apoptosis, and improved cardiac function, which might lead to the development of

AC

novel and effective therapeutic interventions for type I diabetic cardiomyopathy. In diabetic mice, treatment with miR-144 inhibitor enhanced expression of Nrf2 in hearts, which might contribute to the beneficial effect of miR-144 inhibitor. However, Nrf2 was not the unique target of miR-144. It was reported that miR-144 impaired insulin signaling by inhibiting the expression of insulin receptor substrate 1 in type 2 diabetes mellitus [32]. An agomir of miR-144 accelerated plaque formation through impairing reverse cholesterol transport and promoting pro-inflammatory cytokine production by inhibiting expression of ATP binding cassette transporter A1 (ABCA1) [33]. Silencing miR-144 in mice increased hepatic ABCA1 protein, which in turn enhanced plasma

17

ACCEPTED MANUSCRIPT HDL levels [34]. Thus, both of insulin receptor substrate 1 and ABCA1 could be the candidate for further investigation of effect of miR-144 in diabetic cardiomyopathy.

RI P

T

In this work, although miR-144 modulated post-transcription of Nrf2, when exposed to normal glucose, transfection with miR-144 mimic or inhibitor had no significant

SC

effect on Nrf2 protein expression. The protein levels of Nrf2 were modulated exquisitely in cells [35, 36]. Under normal condition, the effect of miR-144 on Nrf2

MA NU

expression in cardiomyocytes might be neutralized through unknown mechanism. Although miR-144 cannot explain the occurred oxidative stress in STZ-induced diabetic hearts (because miR-144 is actually decreased in the diabetic heart),

ED

therapeutic interventions directed at decreasing miR-144 may help to decrease

PT

oxidative stress and apoptosis in the hearts, and pharmacological targeting miR-144 may represent a promising strategy in the management of type I diabetic

CE

cardiomyopathy.

AC

In conclusion, inhibiting microRNA-144 abated oxidative stress, reduced apoptosis, and improved cardiac function in STZ-induced diabetic mice, possibly via enhancing Nrf2 expression.

Conflict of Interest statement The authors declare that they have no conflict of interest.

Author contributions 1. Conception and design of the experiments: Manli Yu, Yu Liu, Xianxian Zhao

18

ACCEPTED MANUSCRIPT 2. Collection, analysis and interpretation of data: Manli Yu, Yu Liu, Bili Zhang,

AC

CE

PT

ED

MA NU

SC

RI P

3. Drafting the article: Manli Yu, Yu Liu, Xianxian Zhao

T

Yicheng Shi, Ling Cui

19

ACCEPTED MANUSCRIPT References 1. Whiting DR, Guariguata L, Weil C, Shaw J. IDF diabetes atlas: global estimates

RI P

T

of the prevalence of diabetes for 2011 and 2030. Diab Res Clin Prac 2011; 94:311–321.

SC

2. Chavali V, Tyagi SC, Mishra PK. Predictors and prevention of diabetic cardiomyopathy. Diabetes Metab Syndr Obes 2013; 6:151–160.

MA NU

3. Wang J, Wang H, Hao P, Xue L, Wei S, Zhang Y, Chen Y. Inhibition of aldehyde dehydrogenase 2 by oxidative stress is associated with cardiac dysfunction in diabetic rats. Mol Med 2011; 17:172–179.

ED

4. Ho FM, Liu SH, Liau CS, Huang PJ, Lin-Shiau SY. High glucose-induced

PT

apoptosis in human endothelial cells is mediated by sequential activations of c-Jun NH(2)-terminal kinase and caspase-3. Circulation 2000; 101:2618–2624.

CE

5. Engel D, Peshock R, Armstong RC, Sivasubramanian N, Mann DL. Cardiac

AC

myocyte apoptosis provokes adverse cardiac remodeling in transgenic mice with targeted TNF overexpression. Am J Physiol Heart Circ Physiol 2004; 287:H1303–H1311. 6. Li Z, Zhang T, Dai H, Liu G, Wang H, Sun Y, Zhang Y, Ge Z. Involvement of endoplasmic reticulum stress in myocardial apoptosis of streptozocin induced diabetic rats. J Clin Biochem Nutr 2007; 41:58–67. 7. Wold LE, Ren J. Streptozotocin directly impairs cardiac contractile function in isolated ventricular myocytes via a p38 map kinase dependent oxidative stress mechanism. Biochem Biophys Res Commun 2004; 318:1066–1071.

20

ACCEPTED MANUSCRIPT 8. Varga ZV, Giricz Z, Liaudet L, Haskó G, Ferdinandy P, Pacher P. Interplay of oxidative, nitrosative/nitrative stress, inflammation, cell death and autophagy in

RI P

T

diabetic cardiomyopathy. Biochim Biophys Acta 2014; 1852:232-242. 9. Dallak MM, Mikhailidis DP, Haidara MA, Bin-Jaliah IM, Tork OM, Rateb MA,

SC

Yassin HZ, Al-Refaie ZA, Ibrahim IM, Elawa SM, Rashed LA, Afifi NA. Oxidative stress as a common mediator for apoptosis induced-cardiac damage in

MA NU

diabetic rats. Open Cardiovasc Med J 2008; 2:70–78.

10. Cai L, Wang Y, Zhou G, Chen T, Song Y, Li X, Kang YJ. Attenuation by metallothionein of early cardiac cell death via suppression of mitochondrial

ED

oxidative stress results in a prevention of diabetic cardiomyopathy. J Am Coll

PT

Cardiol 2006; 48:1688–1697.

11. Fiordaliso F, Bianchi R, Staszewsky L, Cuccovillo I, Doni M, Laragione T, Salio

CE

M, Savino C, Melucci S, Santangelo F, Scanziani E, Masson S, Ghezzi P, Latini R.

AC

Antioxidant treatment attenuates hyperglycemia-induced cardiomyocyte death in rats. J Mol Cell Cardiol 2004; 37:959–968. 12. Shirpoor A, Salami S, Khadem-Ansari MH, Ilkhanizadeh B, Pakdel FG, Khademvatani

K.

Cardioprotective

effect

of

vitamin

E:

rescues

of

diabetes-induced cardiac malfunction, oxidative stress, and apoptosis in rat. J Diabetes Complications 2009; 23:310–316. 13. Filipowicz

W,

Bhattacharyya

SN,

Sonenberg

N.

Mechanisms

of

post-transcriptional regulation by microRNAs: are the answers in sight? Nat Rev Genet 2008; 9:102–114.

21

ACCEPTED MANUSCRIPT 14. Schlüter K-D, Piper HM. Isolation and culture of adult ventricular cardiomyocytes. In: Dhein S, Mohr F, Delmar M (eds) Practical methods in

RI P

T

cardiovascular research. Springer, Berlin, 2008; pp 557–567.

15. Chen C, Ridzon DA, Broomer AJ, Zhou Z, Lee DH, Nguyen JT, Barbisin M, Xu

SC

NL, Mahuvakar VR, Andersen MR, Lao KQ, Livak KJ, Guegler KJ. Real-time quantification of microRNAs by stem-loop RT-PCR. Nucleic Acids Research

MA NU

2005; 33:e179.

16. Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative CT method. Nat Protocols 2008; 3:1101–1108.

ED

17. Elks CM, Mariappan N, Haque M, Guggilam A, Majid DS, Francis J. Chronic

PT

NF-kB blockade reduces cytosolic and mitochondrial oxidative stress and attenuates renal injury and hypertension in SHR. Am J Physiol Renal Physiol

CE

2009; 296:F298–305.

AC

18. Wilson KD, Li Z, Wagner R, Yue P, Tsao P, Nestorova G, Huang M, Hirschberg DL, Yock PG, Quertermous T, Wu JC. Transcriptome alteration in the diabetic heart by rosiglitazone: implications for cardiovascular mortality. PLoS One 2008; 3:e2609. 19. Peng T, Shen E, Fan J, Zhang Y, Arnold JM, Feng Q. Disruption of phospholipase C gamma1 signalling attenuates cardiac tumor necrosis factor-alpha expression and improves myocardial function during endotoxemia. Cardiovasc Res 2008; 78:90–97. 20. Shen E, Li Y, Shan L, Zhu H, Feng Q, Arnold JM, Peng T. Rac1 is required for

22

ACCEPTED MANUSCRIPT cardiomyocyte apoptosis during hyperglycemia. Diabetes 2009; 58:2386–2395 21. Sangokoya C, Telen MJ, Chi JT. microRNA miR-144 modulates oxidative stress

RI P

T

tolerance and associates with anemia severity in sickle cell disease. Blood 2010; 116:4338–4348.

SC

22. Csiszar A, Gautam T, Sosnowska D, Tarantini S, Banki E, Tucsek Z, Toth P, Losonczy G, Koller A, Reglodi D, Giles CB, Wren JD, Sonntag WE, Ungvari Z.

MA NU

Caloric restriction confers persistent anti-oxidative, pro-angiogenic, and anti-inflammatory effects and promotes anti-aging miRNA expression profile in cerebromicrovascular endothelial cells of aged rats. Am J Physiol Heart Circ

ED

Physiol 2014; 307:H292–306.

PT

23. Kumar S, Prasad S, Sitasawad SL. Multiple antioxidants improve cardiac complications and inhibit cardiac cell death in streptozotocin-induced diabetic

CE

rats. PLoS One 2013 8:e67009.

AC

24. Okatan EN, Tuncay E, Turan B. Cardioprotective effect of selenium via modulation of cardiac ryanodine receptor calcium release channels in diabetic rat cardiomyocytes

through

thioredoxin

system.

J

Nutr

Biochem

2013;

24:2110-2118. 25. Giacco F, Brownlee M. Oxidative stress and diabetic complications. Circ Res 2010; 107:1058–1070. 26. Kensler TW, Wakabayashi N, Biswal S. Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu Rev Pharmacol Toxicol 2007; 47:89–116.

23

ACCEPTED MANUSCRIPT 27. Li J, Johnson D, Calkins M,Wright L, Svendsen C, Johnson J. Stabilization of

RI P

human neural stem cells. Toxicol Sci 2005; 83:313–328.

T

Nrf2 by tBHQ confers protection against oxidative stress-induced cell death in

28. Lee JM, Calkins MJ, Chan K, KanYW, Johnson JA. Identification of the

SC

NF-E2-related factor-2-dependent genes conferring protection against oxidative stress in primary cortical astrocytes using oligonucleotide microarray analysis. J

MA NU

Biol Chem 2003; 278:12029–12038.

29. He X, Kan H, Cai L, Ma Q. Nrf2 is critical in defense against high glucose induced oxidative damage in cardiomyocytes. J Mol Cell Cardiol 2009;

ED

46:47–58.

gatekeeping

PT

30. He X, Ma Q. Redox regulation by nuclear factor erythroid 2-related factor 2: for

the

basal

and

diabetes-induced

expression

of

CE

thioredoxin-interacting protein. Mol Pharmacol 2012; 82:887–897.

AC

31. Tan Y, Ichikawa T, Li J, Si Q, Yang H, Chen X, Goldblatt CS, Meyer CJ, Li X, Cai L, Cui T. Diabetic downregulation of Nrf2 activity via ERK contributes to oxidative stress-induced insulin resistance in cardiac cells in vitro and in vivo. Diabetes 2011; 60:625–633. 32. Karolina DS, Armugam A, Tavintharan S, Wong MT, Lim SC, Sum CF, Jeyaseelan K. MicroRNA 144 impairs insulin signaling by inhibiting the expression of insulin receptor substrate 1 in type 2 diabetes mellitus. PLoS One 2011; 6:e22839. 33. Hu YW, Hu YR, Zhao JY, Li SF, Ma X, Wu SG, Lu JB, Qiu YR, Sha YH, Wang

24

ACCEPTED MANUSCRIPT YC, Gao JJ, Zheng L, Wang Q. An agomir of miR-144-3p accelerates plaque formation through impairing reverse cholesterol transport and promoting

RI P

T

pro-inflammatory cytokine production. PLoS One 2014; 9:e94997. 34. de Aguiar Vallim TQ, Tarling EJ, Kim T, Civelek M, Baldán Á, Esau C, Edwards

SC

PA. MicroRNA-144 regulates hepatic ATP binding cassette transporter A1 and plasma high-density lipoprotein after activation of the nuclear receptor farnesoid

MA NU

X receptor. Circ Res 2013; 112:1602–1612.

35. Li W, Thakor N, Xu EY, Huang Y, Chen C, Yu R, Holcik M, Kong AN. An internal ribosomal entry site mediates redox-sensitive translation of Nrf2. Nucleic

ED

Acids Res 2010; 38:778-788.

PT

36. van der Velden AW, Thomas AA. The role of the 5' untranslated region of an mRNA in translation regulation during development. Int J Biochem Cell Biol

AC

CE

1999; 31:87-106.

25

ACCEPTED MANUSCRIPT Table 1. Effect of treatment with miR144 antagomir (anti-miR144) on STZ-induced

Con+anti-miR144 STZ

Body weight (g)

32.6±4.8

31.7±5.2

Fasting blood glucose (mM)

5.3±1.2

5.1±1.0

+dF/dt max (g/s)

72.8±9.1

75.5±8.6

-dF/dt min (g/s)

55.2±6.5

54.1±6.2

FS (%)

57.9±5.2

EF (%)

83.5±5.8

RI P

Con

T

cardiac dysfunction in mice. STZ+anti-miR144 22.7±4.1 *

19.1±4.2 *

19.4±3.1 *

SC

21.6±3.6 *

64.7±8.0 #

26.0±4.8 *

51.9±5.7 #

59.5±6.6

40.6±3.5 *

52.8±4.5 #

84.3±6.0

67.9±5.2 *

80.2±4.9 #

MA NU

44.1±7.2 *

ED

Values are means ± SD. n=9-11 in each group; STZ, streptozotocin; FS, Fractional

PT

shortening; EF, Ejection fraction; * P < 0.05 versus control group, # P < 0.05 versus

AC

CE

STZ group.

26

ACCEPTED MANUSCRIPT Figure legend Figure 1. Levels of miR-144 in heart tissues of STZ-induced diabetic mice. Mice

RI P

T

were injected with STZ to induce diabetes. 8 weeks later, hearts were removed for determination of miR-144 levels by RT-PCR method. Values are means ± SD. n=9 in

SC

each group; STZ, streptozotocin; * P < 0.05 versus control group.

MA NU

Figure 2. miR-144 directly targeted Nrf2 gene. Dual luciferase activity assay of cardiomyocytes cotransfected with the plasmid containing the segment of Nrf2 3’UTR for miR-144 or a control oligoribonucleotide showed that miR-144 inhibited

ED

luciferase activity, compared with controls. Mutation of the miR-144 binding site

PT

abolished this effect (A). Levels of miR-144 (B) in cardiomyocytes exposed to high glucose. Western blot analysis of Nrf2 protein levels after transfection with miR-144

CE

mimic (C) or inhibitor (D) in cardiomyocytes exposed to high glucose. Nrf2, nuclear

AC

factor-erythroid 2-related factor 2; NG, normal glucose; HG, high glucose; * P < 0.05 versus NG group, # P < 0.05 versus HG group.

Figure 3. Effect of miR-144 on high glucose-induced ROS formation and apoptosis in cardiomyocytes. Transfection with miR-144 mimic increased ROS formation (A) and apoptosis (B, C) induced by HG exposure in cultured cells, which was reversed by treatment with Dh404 (0.2μM). Transfection with miR-144 inhibitor attenuated ROS formation (D) and apoptosis (E) induced by HG exposure in cultured cells. TUNEL assay for cardiomyocyte apoptosis. Sections were stained for DAPI to identify

27

ACCEPTED MANUSCRIPT cardiomyocyte nuclei (blue signal, left panel) and for TUNEL activity to identify apoptotic cells (FITC-conjugated secondary antibody, green signal, middle panel).

RI P

T

Apoptotic cardiomyocytes are identified in the merged image. ROS, reactive oxygen species; DAPI, 4’,6-diamidino-2-phenylindole fluorescent dye; TUNEL, terminal

SC

deoxynucleotidyl transferase dUTP nick end labeling; NG, normal glucose; HG, high glucose; * P < 0.05 versus NG group, # P < 0.05 versus HG group, ^ P < 0.05 versus

MA NU

HG group transfected with miR-144 mimic.

Figure 4. Effect of treatment with miR-144 antagomir on cardiac oxidative stress

ED

induced by STZ in mice. Mice were injected with STZ to induce diabetes, and treated

PT

with miR-144 antagomir. Left ventricles were removed for determination of Nrf2 protein expression (A), ROS formation (B), and MDA levels (C). Values are means ±

CE

SD. n=9-11 in each group; STZ, streptozotocin; Nrf2, nuclear factor-erythroid

AC

2-related factor 2; ROS, reactive oxygen species; MDA, malondialdehyde; * P < 0.05 versus control group, # P < 0.05 versus STZ group. Figure 5. Effect of treatment with miR-144 antagomir on apoptosis induced by STZ in mice. Mice were injected with STZ to induce diabetes, and treated with miR-144 antagomir. Left ventricles were removed for determination of caspase-3 activation and apoptosis, by Western blot and TUNEL, respectively. Caspase-3 activation was calculated by the ratio of cleaved caspase-3 to Pro-caspase-3. TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling; STZ, streptozotocin; * P < 0.05 versus control group, # P < 0.05 versus STZ group.

28

MA NU

SC

RI P

T

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

Figure 1

29

ED

MA NU

SC

RI P

T

ACCEPTED MANUSCRIPT

AC

CE

PT

Figure 2

30

CE

AC

Figure 3

PT

ED

MA NU

SC

RI P

T

ACCEPTED MANUSCRIPT

31

AC

CE

PT

ED

MA NU

SC

RI P

T

ACCEPTED MANUSCRIPT

Figure 4

32

PT

ED

MA NU

SC

RI P

T

ACCEPTED MANUSCRIPT

AC

CE

Figure 5

33

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

MA NU

SC

RI P

T

Funding: This study was supported by the National Natural Science Foundation of China (81170223, 81400287) and Natural Science Foundation of Shanghai (81170223). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

34

Inhibiting microRNA-144 abates oxidative stress and reduces apoptosis in hearts of streptozotocin-induced diabetic mice.

Hyperglycemia-induced reactive oxygen species (ROS) generation contributes to the development of diabetic cardiomyopathy. However, little is known abo...
805KB Sizes 0 Downloads 11 Views