• Ei tuloksia

Functional and biochemical responses of skeletal muscle following a moderate degree of systemic iron loading in mice

N/A
N/A
Info
Lataa
Protected

Academic year: 2022

Jaa "Functional and biochemical responses of skeletal muscle following a moderate degree of systemic iron loading in mice"

Copied!
38
0
0

Kokoteksti

(1)

UEF//eRepository

DSpace https://erepo.uef.fi

Rinnakkaistallenteet Terveystieteiden tiedekunta

2019

Functional and biochemical responses of skeletal muscle following a

moderate degree of systemic iron loading in mice

Liang, C

American Physiological Society

Tieteelliset aikakauslehtiartikkelit

© The American Physiological Society All rights reserved

http://dx.doi.org/10.1152/japplphysiol.00237.2018

https://erepo.uef.fi/handle/123456789/7557

Downloaded from University of Eastern Finland's eRepository

(2)

Original Article

1

2

Functional and biochemical responses of skeletal muscle following a moderate degree of 3

systemic iron loading in mice

4

5

Chen Liang1, Marisa C. Mickey1, Candace N. Receno1, Mustafa Atalay2, and Keith C. DeRuisseau1

6

7

1Department of Exercise Science, Syracuse University, 820 Comstock Ave, Room 201 WB,

8

Syracuse, NY 13244, USA

9

2Institute of Biomedicine, Physiology, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio,

10

Finland

11

12

Author Contributions:

13

C.L., M.A., and K.C.D. conceived and designed research; C.L., M.C.M, and C.N.R. performed

14

experiments; C.L. and K.C.D. analyzed data; C.L., M.A., and K.C.D interpreted results of experiments;

15

C.L. and K.C.D prepared figures; C.L. and K.C.D drafted the manuscript; C.L., C.N.R., M.C.M, M.A.,

16

and K.C.D. edited and revised the manuscript; C.L., M.C.M, C.N.R., M.A., and K.C.D. approved the final

17

version of the manuscript.

18 19

Running Head: Moderate iron loading effects on skeletal muscle

20

Key Words: oxidative stress; non-heme iron; glutathione; thioredoxin; proteolysis

21

Number of Words: 5378

22

Total Number of References: 52

23

24

Corresponding Author:

25

(3)

Keith C. DeRuisseau, Ph.D.

26

Syracuse University

27

Department of Exercise Science

28

820 Comstock Ave, Room 201 WB

29

Syracuse, NY 13244

30

Tel: 315-443-9698

31

Email: kcderuis@syr.edu

32

33 34 35 36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

51

52

(4)

ABSTRACT

53

Excessive iron loading may cause skeletal muscle atrophy and weakness due to its free radical generating

54

properties. To determine whether a clinically relevant degree of iron loading impairs skeletal muscle

55

function, young male mice received injections (i.p.) of iron dextran (4 mg iron/200 µL) or 2 mM D-

56

glucose (control) 5 days/week for two weeks (n = 10/group). Systemic iron loading induced an

57

approximate 4-fold increase in the skeletal muscle non-heme iron (NHI) concentration. Soleus specific

58

tension (1, 30-250 Hz) was lower among iron-loaded animals compared to controls despite similar body

59

mass and muscle mass. Soleus lipid peroxidation (4-hydroxynonenal adducts) and protein oxidation

60

(protein carbonyls) levels were similar between groups. In gastrocnemius muscle, reduced glutathione

61

(GSH) and glutathione peroxidase (GPX) activity were similar but glutathione disulfide (GSSG) and

62

GSSG/GSH ratio were greater in iron-loaded muscle. A greater protein expression level of endogenous

63

thiol antioxidant thioredoxin (TRX) was observed among iron-loaded muscle while its endogenous

64

inhibitor thioredoxin-interacting protein (TXNip) and TRX/TXNip ratio were similar. Glutaredoxin2

65

(GRX2), a thiol-disulfide oxidoreductase activated by GSSG-induced destabilization of its iron-sulfur

66

[2Fe-2S] cluster, was lower following iron loading. Additionally, protein levels of α-actinin and αII-

67

spectrin at 240 kDa were lower in the iron-loaded group. Ryanodine receptor (RyR1) stabilizing subunit

68

calstabin1 was also lower following iron loading. In summary, the contractile dysfunction that resulted

69

from moderate iron loading may be mediated by a disturbance in the muscle redox balance and from

70

changes arising from an increased proteolytic response and aberrant sarcoplasmic reticulum Ca2+ release.

71

72

73

74

75

76

77

78

(5)

NEW & NOTEWORTHY

79

While severe iron loading is known to cause muscle oxidative stress and dysfunction, the effects of a

80

moderate degree of systemic iron loading on muscle contractile function and biochemical responses

81

remain unclear. This study demonstrates that a pathophysiological elevation in the skeletal muscle iron

82

load leads to force deficits that coincide with impaired redox status and structural integrity, and lower

83

ryanodine receptor-associated calstabin1 in the absence of muscle mass changes or oxidative damage.

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

(6)

INTRODUCTION

105

Iron is a trace element well known for its involvement with various physiological processes that

106

include oxygen transport, inflammatory response, energy metabolism, and DNA synthesis (48). While the

107

total body iron is principally localized to the erythron, a sizeable amount of iron (10-15%) is contained

108

within skeletal muscle that is needed for heme and non-heme iron (NHI) containing proteins including

109

myoglobin, cytochromes and other enzymes required for energy metabolism (18). Although the study of

110

skeletal muscle iron metabolism has largely centered on the effects of iron deficiency, recent work is

111

drawing attention to metabolic and functional aberrations that may result from an increased muscle iron

112

status. Notably, associations between an elevated skeletal muscle iron level with conditions including

113

aging-associated muscle atrophy (2, 16, 21, 25, 51, 52), diabetes (22), hemochromatosis (23), and obesity

114

(34) were reported. Moreover, the overall increase in muscle iron under these various conditions ranged

115

between 2 to 7-fold above values observed in healthy controls. Thus, accumulated iron within this range

116

may play an important role in skeletal muscle functional impairment and contribute to aging-associated

117

muscle dysfunction and pathophysiology of certain diseases.

118

While the precise mechanism(s) by which elevated iron imparts toxic effects within skeletal

119

muscle is unknown, it is likely to be mediated via the increased production of reactive oxygen species

120

(ROS) (14). Specifically, iron can exist as low molecular weight complexes (i.e., reactive iron or labile

121

iron) that are capable of generating free radicals via Fenton chemistry (19). Thus, elevated iron levels

122

could contribute to a pathophysiological state in muscle by triggering proteolysis (21, 35) and myonuclear

123

apoptosis (45) by inducing ROS production. For example, iron loading is linked to increased skeletal

124

muscle free radical generation (3), disturbed redox homeostasis (2), and altered cardiac muscle calcium

125

regulation (28, 36), which are factors that can result in reduced muscle contractility. In addition, induction

126

of muscle iron overload in mice via intraperitoneal (i.p.) injection of iron dextran led to elevated ROS

127

production and reductions in muscle mass and strength (39). However, the total muscle iron level

128

achieved in the previous study was much greater than what is typically observed under various

129

(7)

pathological states. Thus, understanding the impact of a moderate degree of iron loading on skeletal

130

muscle function and adaptation responses is clinically relevant.

131

The purpose of this study was to determine how a moderate degree of muscle iron loading within

132

a physiologically relevant range (i.e., increase of 2 to 7-fold) impacts skeletal muscle functional and

133

biochemical responses. It was hypothesized that iron loading would result in muscle contractile

134

dysfunction that would coincide with increased oxidative stress, antioxidant expression and activity, and

135

impaired structural integrity. It was also postulated that iron loading would result in less calstabin1

136

associated with the ryanodine receptor, which would signify dysregulated calcium release from the

137

sarcoplasmic reticulum. Identifying potential consequences of an elevated muscle iron status could

138

provide greater understanding of how clinical disease conditions and aging adversely affect skeletal

139

muscle mass and function.

140 141

METHODS

142

Ethical Approval and Experimental Animals

143

Male CD-1 mice were obtained from Charles River Laboratories. Mice were housed in groups of four and

144

maintained on a 12 hr-12 hr, light-dark cycle under standard laboratory conditions. All mice were

145

provided with autoclaved Purina LabDiet 5010 and water ad libitum. The animal use protocol was

146

approved by the Syracuse University Institutional Animal Care and Use Committee and all experimental

147

procedures followed guidelines established by the American Physiological Society for the use of animals

148

in research.

149 150

Experimental Design

151

Twenty, 12-week-old male mice were randomly divided into iron treatment or control groups (n =

152

10/group). Mice in both groups received 200 μL i.p. injections five days/week for 2 weeks. The iron

153

treatment group received 4 mg of iron dextran/injection (Sigma, St Louis, MO) diluted in saline. The iron

154

loading protocol was designed to increase skeletal muscle iron concentration to a physiologically relevant

155

(8)

level shown to be associated with maladaptive states. Control animals received saline i.p. injections

156

containing 2 mM D-glucose. On the day of tissue collection, animals were anaesthetized by an i.p.

157

injection of Fatal Plus (Vortech Pharmaceuticals Ltd., Dearborn, MI) (80-120 mg/kg). Once in a surgical

158

plane of anesthesia the right soleus muscle was quickly excised and utilized for muscle contractile

159

experiments. Additional muscles including the left soleus, extensor digitorum longus (EDL), and

160

gastrocnemius were harvested and trimmed of excess fat, weighed, frozen in liquid nitrogen and stored at

161

−80°C for biochemical analysis. The liver was also harvested, frozen and stored at −80°C. The mice were

162

euthanized by removal of the heart and diaphragm.

163 164

Soleus Muscle Contractile Properties

165

Following dissection from the animal, the right soleus muscle was immediately transferred to a Krebs-

166

Henseleit buffer containing 12 μM d-tubocurarine (Sigma) and aerated with 95% O2 and 5% CO2 (pH =

167

7.4). The soleus was secured by the tendons using two lightweight plexiglass clamps (Harvard Apparatus,

168

Holliston, MA) and vertically placed between platinum wire stimulating electrodes in an organ bath

169

maintained at 25°C. The distal end of the muscle was secured to a fixed Plexiglas rod. The proximal end

170

of the muscle was connected to a force transducer (300C; Aurora Scientific, Aurora, Ontario, Canada) and

171

the muscle was electrically stimulated to generate a series of twitch forces recorded via a data-acquisition

172

system (Dynamic Muscle Control v4.1.6; Aurora Scientific). The muscle was stimulated with a stimulus

173

duration of 0.5 ms to obtain optimal length (Lo) by adjusting the muscle length using a micrometer. Once

174

Lo was determined the muscle length was measured by a caliper and all force measurements were

175

obtained isometrically at Lo. To determine the force-frequency response, the muscle was stimulated at

176

frequencies of 1, 15, 30, 50, 80, 120, 150, 250, and 300 Hz using a train duration of 500 ms with a 2-min

177

interval between each stimulus. Peak tetanic tension (Po) was identified using a stimulus frequency of 120

178

Hz. Following a 2-min recovery, the muscle underwent a fatigue protocol that consisted of repetitive

179

stimulation (40 Hz, 0.5 train/s, and 300 ms train duration) for 5 min. Force was recorded continuously up

180

to 15 min during the recovery period and data points at 30 s and 1, 2, 5, 10, 15 min were selected to plot

181

(9)

recovery. At the end of the experiment, the muscles were removed, trimmed of excess fat, weighed, and

182

frozen for non-heme iron (NHI) measurement. Muscle force was expressed in absolute, relative (% of

183

peak tetanic tension), and/or values normalized to muscle mass or cross-sectional area, which was

184

calculated by dividing muscle mass by the product of muscle density (1.06 g/cm3) and fiber length (13).

185

The fiber length coefficient of 0.71 was used in the calculation (10).

186 187

Muscle and Liver Non-heme Iron

188

NHI was measured according to the method described by Rebouche et al. (40). Briefly, muscle and liver

189

samples (~10 mg) were homogenized (1:40; wt:vol) on ice in ultrapure water (18.6 MΩ) using a Dounce

190

homogenizer. An equal volume of protein precipitation solution containing 1 N HCL and 10%

191

trichloroacetic acid was added to the tissue homogenates and mixed in 1.5 mL polypropylene tubes.

192

Samples were vortexed and placed on a heat-block at 95°C for 1 hr. Following incubation, the samples

193

were allowed to cool, vortexed, and centrifuged 8,200 x g for 10 min. The supernatant (100 μL) was

194

aliquoted and added to an equal volume of chromagen solution containing 0.508 mM ferrozine, 1.5 M

195

sodium acetate, and 0.1% thioglycolic acid, and blank solution containing 1.5 M sodium acetate, and 0.1%

196

thioglycolic acid. The absorbance was measured at 562 nm (PowerWave HT, BioTek Instruments,

197

Winooski, VT) following a 30-min incubation. NHI concentration was calculated from a standard curve

198

(0, 2, 4, 6, 8, and 10 μg/mL) made by mixing protein precipitation solution with atomic absorption iron

199

standard (Sigma).

200 201

Western Blotting

202

Soleus and gastrocnemius muscles were homogenized (1:20; wt:vol) in RIPA Lysis buffer (Santa Cruz

203

Biotechnology, Dallas, TX) and centrifuged at 10,000 x g for 10 min at 4°C. The protein content of the

204

soluble fraction was assessed by using the RC/DC protein assay (Bio-Rad, Hercules, CA). Proteins (~40

205

μg) were individually separated by SDS-PAGE via 12% polyacrylamide gels containing 0.1% SDS.

206

Following electrophoresis, the proteins were transferred to nitrocellulose membranes using a Mini-

207

(10)

Protean Trans-Blot module (276 mA for 2 hr) or an iBlot system (Invitrogen, Thermo Fisher Scientific,

208

Waltham, MA). The membranes were then blocked in PBS-Tween buffer containing 5.0% skim milk

209

protein and 0.05% Tween-20 and incubated with a primary antibody directed against 4-hydroxynonenal

210

adducts (4-HNE, Alpha Diagnostics, San Antonio, TX,RRID:AB_2629282), thioredoxin (TRX, Cayman

211

Chemical, Ann Arbor, MI, RRID:AB_2725744), thioredoxin-interacting protein (TXNip, MBL

212

international, Woburn, MA, RRID:AB_592934), catalase (CAT, CalBiochem, San Diego, CA,

213

RRID:AB_10726597), copper-zinc superoxide dismutase (Cu-ZnSOD, Novus Biologicals, Littleton, CO,

214

RRID:AB_925658), manganese superoxide dismutase (MnSOD, Cayman Chemical, RRID:AB_1213308),

215

αII-spectrin (Santa Cruz, RRID:AB_671135), glutaredoxin2 (GRX2; MyBiosource, San Diego, CA, Cat#

216

MBS2525062), α-actinin (GeneTex, Irvine, CA, RRID:AB_385929), ryanodine receptor (RyR1; Thermo

217

Fisher Scientific, RRID:AB_2254138), calstabin1 (Thermo Fisher Scientific, RRID:AB_2102731), or

218

GAPDH (Cell Signaling Technology, Danvers, MA, RRID:AB_561053) overnight at 4°C. This step was

219

followed by incubation with a horseradish peroxidase–antibody conjugate (1:1,000-1:5,000) directed

220

against the primary antibody for 1 hr at RT. The membranes were washed and subsequently treated with

221

chemiluminescent reagents (Bio-Rad) and quantified using the ChemiDoc MP imaging system (Bio-Rad).

222 223

Protein Carbonyls

224

Protein carbonyls were measured as a biomarker of protein oxidation by using a commercially available

225

OxyBlot assay kit (Millipore, Billerica, MA). Briefly, muscle samples (~10 mg) were derivatized to 2,4-

226

dinitrophyenylhydrazone (DNP) and neutralized by 2 M Tris base in 30% glycerol. Following

227

electrophoresis using 12% polyacrylamide gels, the proteins were transferred to nitrocellulose membranes.

228

Protein bands were visualized and quantified using the same procedure as described above.

229 230

Immunoprecipitation

231

(11)

Gastrocnemius muscle homogenate was centrifuged (10,000 x g) at 4°C. Supernatants (10 µg/µL) were

232

removed and incubated with protein A/G PLUS-Agarose beads (Santa Cruz) and mouse IgG (Santa Cruz)

233

for 1 hour at 4°C to minimize nonspecific binding. After brief centrifugation the supernatants were

234

removed and the beads were discarded. RyR1 was immunoprecipitated from the pre-cleared supernatants

235

using an anti-RyR1 antibody (6 µg) incubated for 4 h followed by 24 h incubation with protein A/G

236

PLUS-Agarose beads at 4°C. Following centrifugation the supernatant was removed and the beads were

237

washed three times with PBS buffer. After adding Laemmli sample buffer to the beads, the sample was

238

heated and loaded onto SDS/PAGE (4-20% gradient) precast gels (Bio-Rad). Proteins were transferred to

239

nitrocellulose membranes using a Mini-Protean Trans-Blot module (276 mA for 2 hr) and a Western blot

240

was then performed on the membranes using RyR1 and calstabin1 (Thermo Fisher Scientific) primary

241

antibodies as described above. Calstabin1 expression was expressed relative to RyR1 protein expression

242

of the same sample.

243 244

Superoxide Dismutase Activity

245

Total superoxide dismutase (SOD) activity was measured using a commercially available assay kit

246

(Sigma). Muscle homogenate (1:20; wt:vol) was mixed with RIPA Lysis buffer (Santa Cruz) and

247

centrifuged at 10,000 x g for 10 min at 4°C. Supernatant (20 μL) was added to 200 µL of WST Working

248

Solution and 20 µL of Enzyme Working Solution, or 20 µL of Dilution buffer, and incubated at 37°C for

249

20 min. Absorbance were determined at 450 nm by using a spectrometer (PowerWave HT, BioTek

250

Instruments).

251 252

Glutathione Peroxidase Activity

253

Glutathione peroxidase (GPX) activity was measured from muscle homogenate (1:20; wt:vol) with tert-

254

butyl hydroperoxide as substrate mixed with buffer containing 50 mM Tris-HCL and 0.1 mM EDTA in

255

the presence of 1 mM NADPH, 2.5 mM GSH, and 1.5 U/ml GSSG reductase (47). Activity was

256

determined by monitoring the reduction in absorbance per minute according to the oxidation of NADPH

257

(12)

at 340 nm (PowerWave HT, BioTek). Activity was normalized to protein concentration of the sample and

258

expressed as units per mg protein.

259 260

Glutathione

261

Total glutathione (TGSH) was measured using supernatant (10 μL) from muscle homogenate (1:20;

262

wt:vol) centrifuged at 10,000 x g for 5 min at 4°C. Supernatant (20 μL) was diluted 10-fold and reacted

263

with 0.25 mM 5,5′-dithiobis-(2- nitrobenzoic acid), 0.4 mM NADPH, and 0.6875 U/ml glutathione

264

reductase (Sigma). GSSG was determined from muscle homogenate supernatant prepared in 5%

265

metaphosphoric acid, and derivatized with 2% 2-vinylpyridine and triethanolamine. TGSH and GSSG

266

were determined by monitoring the formation of 2-nitro-5-thiobenzoic acid at 412 nm (PowerWave HT,

267

BioTek). The concentrations of TGSH and GSSG were obtained by linear regression from the standard

268

curves and GSH was derived from subtracting GSSG from TGSH.

269 270

Statistical Analyses

271

All measures were analyzed using independent sample t-test to compare group differences. The

272

assumptions of normality and homogeneity of variance were verified using Shapiro-Wilk test and

273

Levene’s test, respectively. Box-Cox transformation was used if the assumption of normality was violated.

274

A repeated-measures analysis of variance (ANOVA) was used to assess for the analysis of contractile

275

function changes across different frequency levels. Mauchly’s criterion was used to determine whether

276

data violated the assumption of homogeneity of variance, and Greenhouse-Geiser corrected significance

277

values are reported alternatively when violation occurred. Test statistics of overall multivariate

278

significance was achieved using Wilk’s Lambda. Area under the curve was calculated for force-frequency,

279

fatigue, and recovery data using the trapezoidal integral method. Data are graphically depicted by boxplot

280

in which whiskers represent the 5th and 95th percentiles and mean represented by a plus sign (+). A

281

significance level of P < 0.05 was selected for all analyses, which were performed using SPSS v. 24.0

282

(IBM Corp., Armonk, NY). Tabular data are reported as mean ± SD.

283

(13)

RESULTS

284

Non-heme Iron Level in Muscle and Liver

285

The NHI level of muscles and liver is reported in Table 1. To ensure that the iron injection protocol

286

resulted in a moderate increase in skeletal muscle iron status, we examined the NHI level of the soleus,

287

EDL, and gastrocnemius muscles, in addition to the liver. NHI level of the muscles and liver from the

288

iron-loaded mice was significantly greater than control (P < 0.01) values. Specifically, 10 days of iron

289

injections resulted in a 3.7 to 4-fold increase in the muscle iron level. The liver NHI level was 5.2-fold

290

greater compared to the value of the control group.

291 292

Body Mass (BM) and Muscle Mass (MM) Characteristics

293

Animal BM and MM values are displayed in Table 2. Body mass (P = 0.40) and soleus muscle mass (P =

294

0.55) were not significantly different between iron and control groups. Soleus MM/BM ratio (P = 0.39)

295

was also not significantly different between groups.

296 297

Soleus Muscle Contractile Properties

298

Soleus muscle twitch and tetanus contractile properties are displayed in Table 3. There was no significant

299

difference in absolute (P = 0.34) or normalized (P = 0.13) twitch force of muscles between iron-loaded

300

mice and controls. Also, no significant difference was observed for time to peak tension (P = 0.16), or

301

half-relaxation (P = 0.21) time between groups. However, when normalized to muscle mass (P = 0.02)

302

and calculated physiological cross-sectional area (P = 0.003), maximal isometric tetanic tension of

303

muscles from iron-loaded animals was significantly lower than controls.

304

To assess whether a moderate increase in muscle iron level compromised muscle function, we

305

also determined the force-frequency response of the soleus muscle (Fig. 1). There was a significant

306

interaction between group and stimulation frequency (P = 0.001). Muscle from iron-loaded mice

307

displayed lower force values at stimulation frequencies tested at 1 Hz and between 30 to 250 Hz (P <

308

(14)

0.05). Also, the area under the curve for normalized tension was significantly lower (P = 0.006) for iron-

309

loaded mice compared with controls.

310

The response of the soleus muscle to fatiguing contractions and recovery is shown in Fig. 2.

311

There was an expected progressive reduction in force generation during the stimulation protocol. No

312

significant difference between groups was observed for area under the curve during fatigue (P = 0.63).

313

Following the fatigue-inducing protocol, muscle force gradually recovered over the 15-min recovery

314

period. The area under the curve during recovery was not significantly different between groups (P =

315

0.74).

316 317

Oxidative Damage and Antioxidant Enzyme Measures

318

Oxidative damage was determined by measurement of 4-HNE adducts and protein carbonyls. Analysis

319

and representative images of oxidative damage levels from gastrocnemius muscle are shown in Fig. 3.

320

Levels of 4-HNE adducts were not significantly different between iron-loaded and control groups (P =

321

0.99). Protein carbonyls were also not different between groups (P = 0.57).

322

Analysis and representative Western blot images of key antioxidant proteins from gastrocnemius

323

are shown in Fig. 4 and Fig. 5. The protein expression level of TRX (P = 0.01) was significantly greater

324

in the iron-loaded group compared with controls. Similar levels of TXNip (P = 0.50) and TRX/TXNip (P

325

= 0.16) were observed between the two groups. However, GRX2 was lower (P = 0.04) in iron-loaded

326

muscle compared with controls. CAT (P = 0.99), Cu-ZnSOD (P = 0.70) and MnSOD (P = 0.47) protein

327

expression levels were similar between iron animals and controls. Total SOD activity was also similar

328

between groups (P = 0.85).

329

Analysis of antioxidant enzymes and activity related to glutathione metabolism are shown in Fig.

330

6. While GSH (P = 0.39) and GPX activity (P = 0.82) were similar between two groups, muscle from

331

iron-loaded mice displayed a significantly greater GSSG (P = 0.03) and GSSG/GSH ratio (P = 0.03).

332 333

Muscle Tissue Structure Integrity

334

(15)

The levels of αII-spectrin and α-actinin were assessed as an indication of myocyte structural integrity. As

335

illustrated in Fig. 7, αII-spectrin at 240 kDa was significantly lower in the iron group compared with

336

controls (P = 0.02). No differences were observed in its cleavage products at 150 kDa (P = 0.41) or 120

337

kDa (P = 0.11). Similar to αII-spectrin, α-actinin levels were also lower in the iron-loaded gastrocnemius

338

muscle (P = 0.04).

339 340

Calcium Release Channel RyR1 and Calstabin1

341

As illustrated in Fig. 8, immunoprecipitation of RyR1-associated calstabin1 was lower in the muscle of

342

iron-loaded mice compared with controls (P = 0.04).

343 344

DISCUSSION

345

The iron loading regimen used in the present study caused a moderate elevation in the muscle NHI level

346

that was associated with diminished muscle force production without changes in muscle mass. The results

347

suggest that oxidative damage may not be a contributing factor to the muscle dysfunction since no

348

changes were observed in lipid peroxidation or protein oxidation measures. However, changes in GRX2

349

and GSH-TRX system in response to iron loading suggests conditions of an unfavorable redox status. The

350

lower expression level of cytoskeletal proteins in iron-loaded muscle may also be reflective of increased

351

proteolysis and reduced muscle structural integrity, which could be an additional factor responsible for

352

reduced force production. A lower expression of RyR1-associated calstabin1 induced by iron loading also

353

suggests aberrant sarcoplasmic reticulum (SR) Ca2+ release. The implications of these findings are

354

discussed in the sections below.

355 356

Muscle Iron Concentration following the 10-day Iron Loading Regimen

357

The degree of iron loading achieved in the muscles was within the range of iron levels reported for

358

various pathological conditions. Huang et al. (22, 23) reported a 2.1-fold greater skeletal muscle NHI and

359

(16)

a 2.3-fold greater ferritin level of Hfe-/- mice, which is a model of hemochromatosis. Rats treated with

360

STZ, a Type-I diabetes animal model, showed a 1.4-fold increase in muscle iron content (43). Using MRI,

361

obese patients exhibited 1.2-fold greater paravertebral muscle iron level than non-obese individuals (34).

362

Moreover, our group previously reported a 2 to 7-fold greater NHI concentration of the plantaris over the

363

progression of aging in rats (16). Our current findings are consistent with those of others (51) whereby

364

gastrocnemius muscle iron concentration was 3.4-fold higher in 32-month-old rats as compared with

365

young animals. Thus, the moderate increase in muscle iron level achieved in this study is likely to be

366

reflective of states that are clinically relevant.

367 368

A high degree of muscle iron loading was previously shown to reduce endurance capacity and muscle

369

strength in mice (39). However, the high degree of muscle iron loading that was achieved in the previous

370

study appears to be beyond what is typically observed in pathological conditions. During our data

371

collection, a study was published that examined cell signaling and muscle atrophy responses to moderate

372

iron loading (24). Daily iron injections (10 mg iron dextran) for one week resulted in a 2.0-fold greater

373

gastrocnemius iron level compared with controls. Notably, iron-loaded muscle displayed atrophy and

374

reduced phosphorylation of Akt-forkhead box O3 signaling. Since these findings were divergent to our

375

current data we conducted a pilot experiment to closely replicate part of the experimental design. Briefly,

376

12-week-old male CD-1 mice were randomly divided into control (2 mM D-glucose in saline i.p.; n = 4)

377

and 10 mg iron dextran (i.p.; n = 5) groups. All mice received a daily injection for seven consecutive days

378

and gastrocnemius muscles were harvested 24-hrs following the last injection. In contrast to results of the

379

previous report, we observed muscle NHI concentration of mice receiving 10 mg iron to be 5.1-fold

380

greater than controls, which was greater than the iron level achieved by injecting 4 mg of iron for 10-days

381

based on results of this current study. Thus, differences in the methodology used to assess tissue iron may

382

explain why our findings differ from those of Ikeda et al. (24).

383 384

Iron Loading Effects on Muscle Contractile Properties

385

(17)

Muscles from iron-loaded mice exhibited reduced force production. A negative correlation between

386

maximal specific tension and soleus NHI concentration (r = -0.807; P < 0.001) was also observed;

387

indicating a strong link between excessive muscle iron and impaired maximal force generation. The

388

reduction in normalized force values across a broad range of stimulation frequencies occurred in the

389

absence of differences in muscle mass. Our data are consistent with previous data obtained in vivo, in

390

which iron overloaded mice generated lower force on a maximal strength test. Furthermore, the

391

progressive loss of force over repeated trials was greater among the iron overloaded animals (39).

392 393

We postulate that the lower force production of iron-loaded mice may have partially been the result of a

394

deterioration in myofibril structural proteins. This is supported by our result of αII-spectrin and α-actinin

395

showing lower expression in the iron-loaded muscle. Notably, these cytoplasmic proteins can be cleaved

396

by calpains, which are Ca2+ dependent non-lysosomal cysteine proteases. αII-spectrin is located within

397

the contractile fibers close to the SR as well as on the plasma membrane in cardiomyocytes (4) and

398

functions in the maintenance of muscle cell structures and force transmission (5). α-actinin belongs to the

399

family of focal adhesion proteins, linking integrin receptors to the actin cytoskeleton. Since integrins are

400

important sites for force transmission through focal adhesions, the lower expression of α-actinin may be a

401

contributing factor to muscle dysfunction (27). Altered release of Ca2+ from the SR could also play a role

402

in mediating impaired muscle force production and structural integrity. Notably, RyR1 channel-

403

stabilizing subunit calstabin1 was lower in the iron-loaded muscles, which suggests elevated iron may

404

lead to increased calcium leak that could disrupt Ca2+ release during stimulated contractions. Down-

405

regulation of calstabin1 and abnormal Ca2+ leak from the SR was reported to contribute to diaphragm

406

muscle weakness following long-term mechanical ventilation (33). Muscle iron loading may have altered

407

β-adrenergic signaling that could explain the down-regulation of calstabin1 (31, 33). Further study would

408

be necessary to clarify the role of the sympathetic nervous system in Ca2+-relevant mechanism(s) of

409

skeletal muscle functional impairment resulting from an elevated iron load.

410

(18)

Iron-loaded muscle did not exhibit greater fatigability as hypothesized, nor did it show muscle weakness

411

during the post-fatigue period. This finding conflicts with studies showing increased fatigue and lower

412

endurance capacity among hemochromatosis patients (1, 15). The lack of difference in the fatigue

413

response could be a result of testing the muscle at a temperature lower than that of in vivo conditions.

414

Muscles display greater resistance to fatigue at a lower temperature (41) due to lower ROS production. In

415

light of the lower force production observed in the unfatigued state it may be that the experimental

416

condition led to an underestimation of the impact of iron loading on force generation during fatiguing

417

contractions.

418 419

Iron Loading Altered the Muscle Redox Status, but not Markers of Oxidative Damage or

420

Enzymatic Antioxidant Status

421

It was anticipated that iron loading would result in muscle oxidative damage as previous studies showed

422

an association between iron accumulation and increased ROS production. However, we did not observe

423

greater levels of 4-HNE adducts or protein carbonyls in soleus muscle of iron-loaded mice. This could be

424

explained by their increased removal since the accumulation of oxidatively damaged proteins induces

425

proteolytic activity which in turn facilitates damaged protein catabolism (37). Additional tissue iron could

426

also be stored in ferritin, which is protective against oxidative stress since ferritin protein is able to

427

sequester and stabilize iron. However, the dynamics of iron storage/release from ferritin could still lead to

428

increases in labile iron capable of participating in redox reactions (50). Oxidants including superoxide (8,

429

9, 20) and nitric oxide (42) can release iron from the ferritin molecule in vitro. We also anticipated a

430

greater antioxidant response to counter elevated ROS generation incurred by iron loading. However, iron-

431

loaded animals did not show a greater degree of total SOD activity or a difference in protein expression

432

levels of Cu-ZnSOD, MnSOD, or CAT. In contrast, a thiol antioxidant response was revealed as

433

evidenced by a greater TRX protein expression. Through its disulfide reductase activity the TRX system

434

plays an important role in cellular redox status regulation and antioxidant protection. The response of

435

greater TRX in these mice is consistent with a previous report showing greater TRX protein level and

436

(19)

activity in the brain of female rats that were loaded with iron (12). Indeed, TRX is a stress-inducible thiol-

437

containing endogenous antioxidant and redox-regulator, which was shown to be an indicator of oxidative

438

stress in a variety of disease states. For example, the presence of catalytically active iron and induction of

439

oxidative stress in lung-injury models was accompanied by TRX nuclear translocation and its interaction

440

with other redox-sensitive elements (37). It was also reported that TRX was associated with serum ferritin

441

and further increased by iron infusion in hepatitis C virus-infected patients (26). As an endogenous

442

inhibitor of TRX, TXNip expression was unaffected but inconsistent expression of these reciprocal

443

proteins was previously reported (46). Thus, despite greater TRX protein expression in response to an

444

elevated iron level, the unaltered ratio of TRX/TXNip may have limited the contribution of TRX in

445

regulating thiol homeostasis.

446 447

Oxidized glutathione (GSSG) and its ratio to the reduced form (GSH) were greater in iron-loaded muscle,

448

which further supports the occurrence of a shift in antioxidant pools. The greater level of TRX after iron

449

administration may have been a compensatory response to greater GSSG and GSSG/GSH ratio. Moreover,

450

there is cross-talk between the TRX system and GSH metabolism through disulfide yields (38), and

451

GRXs possibly play an important role in their interactions. GRXs are thiol-disulfide oxidoreductases that

452

belong to the TRX family of proteins that are critically involved in regulating redox and iron homeostasis

453

(6). GRX2 is localized to the mitochondria and can accept electrons from GSH and thioredoxin reductase

454

(17). Moreover, GRX2 is a member of the TRX protein family that possesses an iron-sulfur [2Fe-2S]

455

cluster, which may serve as a redox sensor (30). Elevated GSSG/GSH can disrupt the iron-sulfur cluster

456

of dimeric GRX2, resulting in the formation of enzymatically active, iron free monomer of GRX2 (30).

457

However, lower expression of GRX2 resulting from iron loading may have reduced its involvement in

458

redox and iron status control. For example, experimental inhibition of GRX2 in dopaminergic neurons

459

resulted in altered iron regulation, increased level of mitochondrial iron, and reduced iron-sulfur cluster

460

biogenesis and activities of proteins including mitochondrial complex I, mitochondrial aconitase, and

461

cytosolic aconitase (29). Thus, a decreased capacity for FeS cluster synthesis combined with greater iron

462

(20)

release from disrupted GRX2 [2Fe-2S] clusters may increase intracellular labile iron levels that could

463

alter iron regulation and ROS production (29). Furthermore, mice deficient in GRX2 showed altered

464

release of ROS from mitochondria isolated from liver and cardiac muscle (11). Overall, the iron-loading

465

impact on the muscle redox status may extend to the GRX system, in which lowered GRX2 expression

466

may link elevated muscle iron status with an additional mechanism of altered ROS generation and iron

467

status regulation.

468 469

Our findings support the notion that muscle iron loading may lead to disrupted skeletal muscle Ca2+

470

regulation. In addition to calstabin1 dissociation, Ca2+ channel remodeling including RyR1 thio-

471

nitrosylation, oxidation, and Ser-2844 phosphorylation was observed in controlled mechanical

472

ventilation-induced diaphragm muscle weakness (33). Thus, an iron-induced redox disturbance may lead

473

to deregulated Ca2+ homeostasis that activates calpain and triggers degradation of cytoskeletal proteins.

474

Moreover, evidence linking iron associated redox balance disturbances with damage to structural proteins

475

has also been documented. For example, an association between TRX and spectrin was reported in

476

HbEβ-thalassemic erythrocytes whereby upregulated TRX corresponded with disrupted cytoskeleton

477

integrity shown by presence of low molecular weight fragments of β-spectrin (7). A greater GSSG/GSH

478

ratio and modification of the spectrin cytoskeleton network was also displayed among patients with

479

Fanconi's anemia (32). In sickle cells and hippocampal neurons, a connection between a high GSSG/GSH

480

ratio and spectrin ubiquitination was reported. Although in this previous report a high GSSG/GSH ratio

481

led to a reduced level of spectrin ubiquitination and turnover, it provides additional support linking altered

482

redox status with the dysregulation of proteolytic capacity (44). Though there is limited evidence of a

483

direct link of α-actinin and the GSH-TRX system, a study showed that thioredoxin reductase facilitates

484

actin-denitrosylation and β2 integrin-specific adherence through focal adhesion kinase in neutrophils (49).

485

Additional work to directly connect iron loading effects on muscle contractile dysfunction by a

486

mechanism involving redox imbalance and membrane cytoskeleton deterioration is warranted.

487

488

(21)

Summary and Conclusion

489

This study examined skeletal muscle contractile properties in vitro using a mouse model subjected to a

490

moderate degree of systemic iron loading. Although iron loading did not trigger an atrophic response of

491

the muscles, our data indicate that iron may play an important role in the regulation of muscle functional

492

properties. The contractile dysfunction induced by iron may be mediated by perturbations in redox

493

signaling that is part of a regulatory mechanism that leads to an increased proteolytic response and

494

abnormal SR Ca2+ release. Furthermore, iron loading triggers a thiol antioxidant response that may further

495

alter iron status regulation. Future work could further explore thiol antioxidant interactions and

496

implications of elevated skeletal muscle iron status under various pathophysiological conditions.

497 498

Acknowledgements

499

Funding support for these experiments was provided by the Syracuse University SOE.

500 501

Conflict of Interest

502

The authors declare that they have no conflict of interest.

503

504 505

506 507

508 509

510 511

512 513

514 515

516 517

518 519

520 521

522 523

524

(22)

525 526

Reference List

527 528

1. Allen MK. Hereditary hemochromatosis: a literature review and case report. Physiother 529

Can 62: 276-284, 2010.

530

2. Altun M, Edstrom E, Spooner E, Flores-Moralez A, Bergman E, Tollet-Egnell P, 531

Norstedt G, Kessler BM, and Ulfhake B. Iron load and redox stress in skeletal muscle of aged 532

rats. Muscle Nerve 36: 223-233, 2007.

533

3. Bartfay WJ, Dawood F, Wen WH, Lehotay DC, Hou D, Bartfay E, Luo X, Backx 534

PH, and Liu PP. Cardiac function and cytotoxic aldehyde production in a murine model of 535

chronic iron-overload. Cardiovasc Res 43: 892-900, 1999.

536

4. Bennett PM, Baines AJ, Lecomte MC, Maggs AM, and Pinder JC. Not just a plasma 537

membrane protein: in cardiac muscle cells alpha-II spectrin also shows a close association with 538

myofibrils. J Muscle Res Cell Motil 25: 119-126, 2004.

539

5. Bennett V, and Baines AJ. Spectrin and ankyrin-based pathways: metazoan inventions 540

for integrating cells into tissues. Physiol Rev 81: 1353-1392, 2001.

541

6. Berndt C, and Lillig CH. Glutathione, Glutaredoxins, and Iron. Antioxid Redox Signal 542

27: 1235-1251, 2017.

543

7. Bhattacharya D, Saha S, Basu S, Chakravarty S, Chakravarty A, Banerjee D, and 544

Chakrabarti A. Differential regulation of redox proteins and chaperones in HbEbeta- 545

thalassemia erythrocyte proteome. Proteomics Clin Appl 4: 480-488, 2010.

546

8. Bolann BJ, and Ulvik RJ. On the limited ability of superoxide to release iron from 547

ferritin. Eur J Biochem 193: 899-904, 1990.

548

9. Bolann BJ, and Ulvik RJ. Release of iron from ferritin by xanthine oxidase. Role of the 549

superoxide radical. Biochem J 243: 55-59, 1987.

550

10. Brooks SV, and Faulkner JA. Contractile properties of skeletal muscles from young, 551

adult and aged mice. J Physiol 404: 71-82, 1988.

552

11. Chalker J, Gardiner D, Kuksal N, and Mailloux RJ. Characterization of the impact of 553

glutaredoxin-2 (GRX2) deficiency on superoxide/hydrogen peroxide release from cardiac and 554

liver mitochondria. Redox Biol 15: 216-227, 2018.

555

12. Chen TY, Tsai KL, Lee TY, Chiueh CC, Lee WS, and Hsu C. Sex-specific role of 556

thioredoxin in neuroprotection against iron-induced brain injury conferred by estradiol. Stroke 41:

557

160-165, 2010.

558

13. Close RI. Dynamic properties of mammalian skeletal muscles. Physiol Rev 52: 129-197, 559

1972.

560

14. Comporti M. Introduction-serial review: iron and cellular redox status. Free Radic Biol 561

Med 32: 565-567, 2002.

562

15. Davidsen ES, Liseth K, Omvik P, Hervig T, and Gerdts E. Reduced exercise capacity 563

in genetic haemochromatosis. Eur J Cardiovasc Prev Rehabil 14: 470-475, 2007.

564

16. DeRuisseau KC, Park YM, DeRuisseau LR, Cowley PM, Fazen CH, and Doyle RP.

565

Aging-related changes in the iron status of skeletal muscle. Exp Gerontol 48: 1294-1302, 2013.

566

17. Fernando MR, Lechner JM, Lofgren S, Gladyshev VN, and Lou MF. Mitochondrial 567

thioltransferase (glutaredoxin 2) has GSH-dependent and thioredoxin reductase-dependent 568

peroxidase activities in vitro and in lens epithelial cells. FASEB J 20: 2645-2647, 2006.

569

18. Finch CA, and Huebers H. Perspectives in iron metabolism. N Engl J Med 306: 1520- 570

1528, 1982.

571

(23)

19. Gutteridge JM. Iron and oxygen: a biologically damaging mixture. Acta Paediatr Scand 572

Suppl 361: 78-85, 1989.

573

20. Harris LR, Cake MH, and Macey DJ. Iron release from ferritin and its sensitivity to 574

superoxide ions differs among vertebrates. Biochem J 301 ( Pt 2): 385-389, 1994.

575

21. Hofer T, Marzetti E, Xu J, Seo AY, Gulec S, Knutson MD, Leeuwenburgh C, and 576

Dupont-Versteegden EE. Increased iron content and RNA oxidative damage in skeletal muscle 577

with aging and disuse atrophy. Exp Gerontol 43: 563-570, 2008.

578

22. Huang J, Gabrielsen JS, Cooksey RC, Luo B, Boros LG, Jones DL, Jouihan HA, 579

Soesanto Y, Knecht L, Hazel MW, Kushner JP, and McClain DA. Increased glucose disposal 580

and AMP-dependent kinase signaling in a mouse model of hemochromatosis. J Biol Chem 282:

581

37501-37507, 2007.

582

23. Huang J, Jones D, Luo B, Sanderson M, Soto J, Abel ED, Cooksey RC, and 583

McClain DA. Iron overload and diabetes risk: a shift from glucose to Fatty Acid oxidation and 584

increased hepatic glucose production in a mouse model of hereditary hemochromatosis. Diabetes 585

60: 80-87, 2011.

586

24. Ikeda Y, Imao M, Satoh A, Watanabe H, Hamano H, Horinouchi Y, Izawa-Ishizawa 587

Y, Kihira Y, Miyamoto L, Ishizawa K, Tsuchiya K, and Tamaki T. Iron-induced skeletal 588

muscle atrophy involves an Akt-forkhead box O3-E3 ubiquitin ligase-dependent pathway. J 589

Trace Elem Med Biol 35: 66-76, 2016.

590

25. Jung SH, DeRuisseau LR, Kavazis AN, and DeRuisseau KC. Plantaris muscle of aged 591

rats demonstrates iron accumulation and altered expression of iron regulation proteins. Exp 592

Physiol 93: 407-414, 2008.

593

26. Kato A, Odamaki M, Nakamura H, Yodoi J, and Hishida A. Elevation of blood 594

thioredoxin in hemodialysis patients with hepatitis C virus infection. Kidney Int 63: 2262-2268, 595

2003.

596

27. Katsumi A, Orr AW, Tzima E, and Schwartz MA. Integrins in mechanotransduction.

597

J Biol Chem 279: 12001-12004, 2004.

598

28. Kim E, Giri SN, and Pessah IN. Iron(II) is a modulator of ryanodine-sensitive calcium 599

channels of cardiac muscle sarcoplasmic reticulum. Toxicol Appl Pharmacol 130: 57-66, 1995.

600

29. Lee DW, Kaur D, Chinta SJ, Rajagopalan S, and Andersen JK. A disruption in iron- 601

sulfur center biogenesis via inhibition of mitochondrial dithiol glutaredoxin 2 may contribute to 602

mitochondrial and cellular iron dysregulation in mammalian glutathione-depleted dopaminergic 603

cells: implications for Parkinson's disease. Antioxid Redox Signal 11: 2083-2094, 2009.

604

30. Lillig CH, Berndt C, Vergnolle O, Lonn ME, Hudemann C, Bill E, and Holmgren A.

605

Characterization of human glutaredoxin 2 as iron-sulfur protein: a possible role as redox sensor.

606

Proc Natl Acad Sci U S A 102: 8168-8173, 2005.

607

31. Link G, Athias P, Grynberg A, Hershko C, and Pinson A. Iron loading modifies beta- 608

adrenergic responsiveness in cultured ventricular myocytes. Cardioscience 2: 185-188, 1991.

609

32. Malorni W, Straface E, Pagano G, Monti D, Zatterale A, Del Principe D, Deeva IB, 610

Franceschi C, Masella R, and Korkina LG. Cytoskeleton alterations of erythrocytes from 611

patients with Fanconi's anemia. FEBS Lett 468: 125-128, 2000.

612

33. Matecki S, Dridi H, Jung B, Saint N, Reiken SR, Scheuermann V, Mrozek S, 613

Santulli G, Umanskaya A, Petrof BJ, Jaber S, Marks AR, and Lacampagne A. Leaky 614

ryanodine receptors contribute to diaphragmatic weakness during mechanical ventilation. Proc 615

Natl Acad Sci U S A 113: 9069-9074, 2016.

616

(24)

34. Moreno-Navarrete JM, Blasco G, Xifra G, Karczewska-Kupczewska M, 617

Stefanowicz M, Matulewicz N, Puig J, Ortega F, Ricart W, Straczkowski M, and 618

Fernandez-Real JM. Obesity Is Associated With Gene Expression and Imaging Markers of Iron 619

Accumulation in Skeletal Muscle. J Clin Endocrinol Metab 101: 1282-1289, 2016.

620

35. Nagasawa T, Hatayama T, Watanabe Y, Tanaka M, Niisato Y, and Kitts DD. Free 621

radical-mediated effects on skeletal muscle protein in rats treated with Fe-nitrilotriacetate.

622

Biochem Biophys Res Commun 231: 37-41, 1997.

623

36. Nakamura TY, Goda K, Okamoto T, Kishi T, Nakamura T, and Goshima K.

624

Contractile and morphological impairment of cultured fetal mouse myocytes induced by oxygen 625

radicals and oxidants. Correlation with intracellular Ca2+ concentration. Circ Res 73: 758-770, 626

1993.

627

37. Radak Z, Sasvari M, Nyakas C, Pucsok J, Nakamoto H, and Goto S. Exercise 628

preconditioning against hydrogen peroxide-induced oxidative damage in proteins of rat 629

myocardium. Arch Biochem Biophys 376: 248-251, 2000.

630

38. Radak Z, Zhao Z, Koltai E, Ohno H, and Atalay M. Oxygen consumption and usage 631

during physical exercise: the balance between oxidative stress and ROS-dependent adaptive 632

signaling. Antioxid Redox Signal 18: 1208-1246, 2013.

633

39. Reardon TF, and Allen DG. Iron injections in mice increase skeletal muscle iron 634

content, induce oxidative stress and reduce exercise performance. Exp Physiol 94: 720-730, 2009.

635

40. Rebouche CJ, Wilcox CL, and Widness JA. Microanalysis of non-heme iron in animal 636

tissues. J Biochem Biophys Methods 58: 239-251, 2004.

637

41. Reid MB. Free radicals and muscle fatigue: Of ROS, canaries, and the IOC. Free Radic 638

Biol Med 44: 169-179, 2008.

639

42. Reif DW, and Simmons RD. Nitric oxide mediates iron release from ferritin. Arch 640

Biochem Biophys 283: 537-541, 1990.

641

43. Sanchez-Gonzalez C, Lopez-Chaves C, Trenzado CE, Aranda P, Lopez-Jurado M, 642

Gomez-Aracena J, Montes-Bayon M, Sanz-Medel A, and Llopis J. Changes in iron 643

metabolism and oxidative status in STZ-induced diabetic rats treated with bis(maltolato) 644

oxovanadium (IV) as an antidiabetic agent. ScientificWorldJournal 2014: 706074, 2014.

645

44. Sangerman J, Kakhniashvili D, Brown A, Shartava A, and Goodman SR. Spectrin 646

ubiquitination and oxidative stress: potential roles in blood and neurological disorders. Cell Mol 647

Biol Lett 6: 607-636, 2001.

648

45. Seo AY, Xu J, Servais S, Hofer T, Marzetti E, Wohlgemuth SE, Knutson MD, 649

Chung HY, and Leeuwenburgh C. Mitochondrial iron accumulation with age and functional 650

consequences. Aging Cell 7: 706-716, 2008.

651

46. Son A, Nakamura H, Okuyama H, Oka S, Yoshihara E, Liu W, Matsuo Y, Kondo N, 652

Masutani H, Ishii Y, Iyoda T, Inaba K, and Yodoi J. Dendritic cells derived from TBP-2- 653

deficient mice are defective in inducing T cell responses. Eur J Immunol 38: 1358-1367, 2008.

654

47. Tappel AL. Glutathione peroxidase and hydroperoxides. Methods Enzymol 52: 506-513, 655

1978.

656

48. Templeton DM, and Liu Y. Genetic regulation of cell function in response to iron 657

overload or chelation. Biochim Biophys Acta 1619: 113-124, 2003.

658

49. Thom SR, Bhopale VM, Milovanova TN, Yang M, and Bogush M. Thioredoxin 659

reductase linked to cytoskeleton by focal adhesion kinase reverses actin S-nitrosylation and 660

restores neutrophil beta(2) integrin function. J Biol Chem 287: 30346-30357, 2012.

661

(25)

50. Watt GD, Jacobs D, and Frankel RB. Redox reactivity of bacterial and mammalian 662

ferritin: is reductant entry into the ferritin interior a necessary step for iron release? Proc Natl 663

Acad Sci U S A 85: 7457-7461, 1988.

664

51. Xu J, Hwang JC, Lees HA, Wohlgemuth SE, Knutson MD, Judge AR, Dupont- 665

Versteegden EE, Marzetti E, and Leeuwenburgh C. Long-term perturbation of muscle iron 666

homeostasis following hindlimb suspension in old rats is associated with high levels of oxidative 667

stress and impaired recovery from atrophy. Exp Gerontol 47: 100-108, 2012.

668

52. Xu J, Knutson MD, Carter CS, and Leeuwenburgh C. Iron accumulation with age, 669

oxidative stress and functional decline. PLoS One 3: e2865, 2008.

670 671

672

673

674 675

676 677

678 679

680 681

682 683

684 685

686 687

688 689

690 691

692 693

694 695

696 697

698 699

700 701

702 703

704 705

706 707

708

(26)

Table 1. Tissue NHI level (nmol/gww) and mean ratio

709 710

Control Iron Mean Ratio

n 10 10

Soleus 700.8 ± 202.7 2619.7 ± 663.0** 3.7

EDL 383.6 ± 119.8 1547.9 ± 392.5** 4.0

Gastrocnemius 702.5 ± 351.0 2850.3 ± 937.7** 3.7

Liver 1509.1 ± 376.7 7826.3 ± 261.7** 5.2

Values are means ± SD. n = number of animals. Mean Ratio = Mean NHI of the Iron group relative to the Control group. **Different from Control: P < 0.01.

711 712

713 714 715 716 717 718 719 720 721

722

723

724

725

726

727

728

729

730

731

732

733

734

(27)

Table 2. Body mass and muscle mass

735

n BM, g Soleus mass, mg MM/BM Ratio

Control 10 40.3 ± 1.7 9.4 ± 1.0 0.233 ± 0.021

Iron 10 39.5 ± 2.3 9.9 ± 2.2 0.250 ± 0.055

Values are means ± SD. n = number of animals. BM, body mass; MM, muscle mass.

736

737 738

739 740

741 742

743 744

745 746

747 748

749 750

751 752

753 754

755 756

757 758

759

760

761

762

763

764

765

766

767

768

769

770

771

772

773

774

775

(28)

Table 3. Soleus contractile properties

776

Control Iron

n 7 6

Twitch

Pt, mN 42.5 ± 5.4 37.4 ± 11.0

Pt, N/cm2 4.6 ± 0.7 4.0 ± 0.7

TPT, ms 34.3 ± 5.3 38.3 ± 4.1

1/2RT, ms 50.7 ± 8.4 56.7 ± 7.5

Tetanus

Po, mN 240.0 ± 47.0 222.8 ± 62.7 Po/pCSA, N/cm2 26.2 ± 2.5 22.4 ± 0.3*

Po/MM, mN/mg 25.3 ± 1.8 22.4 ± 1.3*

Pt/Po 0.171 ± 0.021 0.178 ± 0.031 Values are means ± SD. n = number of animals. Pt, peak twitch tension; TPT, time to peak twitch tension; 1∕2RT, half-relaxation time;

Po, maximal isometric tetanic tension; pCSA, calculated physiological cross-sectional area; Pt/Po, twitch-to-tetanus ratio. *Different from Control: P < 0.05.

777 778 779

780

781

782

783

784

785

786

787

788

789

790

791

792

(29)

Figure captions

793

Fig. 1. Force frequency response of the soleus muscle obtained from control (n = 7), and iron-loaded (n =

794

8) mice. Values are means ± SD. *Different from Control, P < 0.05.

795 796

Fig. 2. Fatigue and recovery response of soleus muscle. A: Absolute active force during fatiguing

797

contractions and recovery. B: Percentage of initial active force during fatigue and recovery. n = 7 for both

798

control and iron-loaded groups.

799 800

Fig. 3. Oxidative damage in soleus of control and iron-loaded mice. A: Top: Arbitrary optical density of

801

4-HNE adducts expressed relative to GAPDH (n = 9-10 animals/group). Bottom: Representative Western

802

blot image of 4-HNE adducts and GAPDH for one control and iron-loaded muscle, respectively. B: Top:

803

Arbitrary optical density of protein carbonyls (PC) expressed relative to GAPDH (n = 9-10

804

animals/group). Bottom: Representative Western blot image of protein carbonyls and GAPDH for one

805

control and iron-loaded muscle, respectively.

806 807

Fig. 4. Protein levels of TRX system in gastrocnemius of control and iron-loaded mice. A: Top: Arbitrary

808

optical density of TRX expressed relative to GAPDH (n = 9-10 animals/group). Bottom: Representative

809

Western blot image of TRX and GAPDH for one control and iron-loaded muscle, respectively. B: Top:

810

Arbitrary optical density of TXNip expressed relative to GAPDH (n = 9-10 animals/group). Bottom:

811

Representative Western blot image of TXNip and GAPDH for one control and iron-loaded muscle,

812

respectively. C: Arbitrary optical density of TRX/TXNip (n = 9-10 animals/group). D: Top: Arbitrary

813

optical density of GRX2 expressed relative to GAPDH (n = 9-10 animals/group). Bottom: Representative

814

Western blot image of GRX2 and GAPDH for one control and iron-loaded muscle, respectively.

815

*Different from Control, P < 0.05.

816

817

(30)

Fig. 5. Antioxidant enzyme protein expression and activity in gastrocnemius of control and iron-loaded

818

mice. A: Top: Arbitrary optical density of CAT expressed relative to GAPDH (n = 9-10 animals/group).

819

Bottom: Representative Western blot image of CAT and GAPDH for one control and iron-loaded muscle,

820

respectively. B: Top: Arbitrary optical density of Cu-ZnSOD expressed relative to GAPDH (n = 9-10

821

animals/group). Bottom: Representative Western blot image of Cu-ZnSOD and GAPDH for one control

822

and iron-loaded muscle, respectively. C: Top: Arbitrary optical density of MnSOD expressed relative to

823

GAPDH (n = 9-10 animals/group). Bottom: Representative Western blot image of MnSOD and GAPDH

824

for one control and iron-loaded muscle, respectively. D: Total SOD enzyme activity (n = 9-10

825

animals/group).

826 827

Fig. 6. Glutathione and GPX activity in gastrocnemius of control and iron-loaded mice. A: GSH (n = 7-8

828

animals/group). B: GSSG (n = 7-8 animals/group). C: Ratio of GSSG/GSH (n = 7-8 animals/group). D:

829

GPX activity (n = 9-10 animals/group). *Different from Control, P < 0.05.

830 831

Fig. 7. Muscle cytoskeleton protein expression in gastrocnemius of control and iron-loaded mice. A: Top:

832

Arbitrary optical density of αII-spectrin expressed relative to GAPDH (n = 9-10 animals/group). Bottom:

833

Representative Western blot image of αII-spectrin and GAPDH for one control and iron-loaded muscle,

834

respectively. B: Top: Arbitrary optical density of α-actinin expressed relative to GAPDH (n = 9-10

835

animals/group). Bottom: Representative Western blot image of α-actinin and GAPDH for one control and

836

iron-loaded muscle, respectively. *Different from Control, P < 0.05.

837 838

Fig. 8. Immunoprecipitation of calstabin1 in gastrocnemius of control and iron-loaded mice. Top:

839

Arbitrary optical density of calstabin1 expressed relative to immunoprecipitated RyR1 (n = 9-10

840

animals/group). Bottom: Representative Western blot image of calstabin1 and RyR1 for one control and

841

iron-loaded muscle, respectively. *Different from Control, P < 0.05.

842

(31)

10 15 20 25 30

ific Tension (N/cm2)

*

* *

* *

*

(32)

1 2 3 4 5 10 15 20 0.06

0.08 0.10 0.12 0.14 0.16 0.18 0.20 0.22

Time (min)

Force (N)

Control Iron

Fatigue Recovery

40 50 60 70 80 90 100

Percent of initial force (%)

Control Iron

B A

(33)

Control Iron 0.0

0.2 0.4 0.6 0.8

4-HNE Adducts/ GAPDH

4-HNE Adducts

Control Iron

0.0 0.1 0.2 0.3 0.4 0.5

PC/ GAPDH

PC

A B

Viittaukset

LIITTYVÄT TIEDOSTOT

ApoA1 was previously demonstrated to increase cell respiration in skeletal muscle cells (Lehti et al., 2013) and the aim is to determine whether the effects of ApoA1

tieliikenteen ominaiskulutus vuonna 2008 oli melko lähellä vuoden 1995 ta- soa, mutta sen jälkeen kulutus on taantuman myötä hieman kasvanut (esi- merkiksi vähemmän

nustekijänä laskentatoimessaan ja hinnoittelussaan vaihtoehtoisen kustannuksen hintaa (esim. päästöoikeuden myyntihinta markkinoilla), jolloin myös ilmaiseksi saatujen

Ydinvoimateollisuudessa on aina käytetty alihankkijoita ja urakoitsijoita. Esimerkiksi laitosten rakentamisen aikana suuri osa työstä tehdään urakoitsijoiden, erityisesti

Mansikan kauppakestävyyden parantaminen -tutkimushankkeessa kesän 1995 kokeissa erot jäähdytettyjen ja jäähdyttämättömien mansikoiden vaurioitumisessa kuljetusta

Työn merkityksellisyyden rakentamista ohjaa moraalinen kehys; se auttaa ihmistä valitsemaan asioita, joihin hän sitoutuu. Yksilön moraaliseen kehyk- seen voi kytkeytyä

Aineistomme koostuu kolmen suomalaisen leh- den sinkkuutta käsittelevistä jutuista. Nämä leh- det ovat Helsingin Sanomat, Ilta-Sanomat ja Aamulehti. Valitsimme lehdet niiden

Muscle weakness in respiratory and peripheral skeletal muscles in a mouse model for nebulin-based nemaline myopathy. Muscle weakness in a mouse model of nemaline myopathy can