IGF-1 prevents ANG II-induced skeletal muscle atrophy via Akt- and. Foxo-dependent inhibition of the ubiquitin ligase atrogin-1 expression.

Similar documents
FOXO Reporter Kit PI3K/AKT Pathway Cat. #60643

Phospho-AKT Sampler Kit

Endogenous glucocorticoids and impaired insulin signaling are both required to stimulate muscle wasting under pathophysiological conditions in mice

HEK293FT cells were transiently transfected with reporters, N3-ICD construct and

Supplemental Data. TGF-β-mediated mir-181a expression promotes breast cancer metastasis by targeting Bim.

Atrogin-1, a muscle-specific F-box protein highly expressed during muscle atrophy

JunB Transcription Factor Maintains Skeletal Muscle Mass and Promotes Hypertrophy

MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells

SUPPLEMENTARY FIGURES AND TABLE

A Central Role of MG53 in Metabolic Syndrome. and Type-2 Diabetes

p47 negatively regulates IKK activation by inducing the lysosomal degradation of polyubiquitinated NEMO

The functional investigation of the interaction between TATA-associated factor 3 (TAF3) and p53 protein

Soft Agar Assay. For each cell pool, 100,000 cells were resuspended in 0.35% (w/v)

Intolerance in Heart Failure

Growth and Differentiation Phosphorylation Sampler Kit

Protection against doxorubicin-induced myocardial dysfunction in mice by cardiac-specific expression of carboxyl terminus of hsp70-interacting protein

Supplementary Figure 1

Part-4. Cell cycle regulatory protein 5 (Cdk5) A novel target of ERK in Carb induced cell death

Doctor of Philosophy

2.5. AMPK activity

IL-6 and Serum Amyloid A Synergy Mediates Angiotensin II Induced Muscle Wasting

The Regulation of Skeletal Muscle Protein Turnover During the Progression of Cancer Cachexia in the Apc Min/+ Mouse

Supplementary Information Titles Journal: Nature Medicine

Supplementary Figure 1. Confocal immunofluorescence showing mitochondrial translocation of Drp1. Cardiomyocytes treated with H 2 O 2 were prestained

Supplementary Figure 1

A Hepatocyte Growth Factor Receptor (Met) Insulin Receptor hybrid governs hepatic glucose metabolism SUPPLEMENTARY FIGURES, LEGENDS AND METHODS

BIO360 Fall 2013 Quiz 1

Serum Amyloid A3 Gene Expression in Adipocytes is an Indicator. of the Interaction with Macrophages

Sestrin2 and BNIP3 (Bcl-2/adenovirus E1B 19kDa-interacting. protein3) regulate autophagy and mitophagy in renal tubular cells in. acute kidney injury

Cell isolation. Spleen and lymph nodes (axillary, inguinal) were removed from mice

General Laboratory methods Plasma analysis: Gene Expression Analysis: Immunoblot analysis: Immunohistochemistry:

Supplementary Figure 1 IL-27 IL

SUPPLEMENTARY INFORMATION

Supplemental Data. Short Article. ATF4-Mediated Induction of 4E-BP1. Contributes to Pancreatic β Cell Survival. under Endoplasmic Reticulum Stress

Hunk is required for HER2/neu-induced mammary tumorigenesis

A growth stimulus is needed for IGF-1 to induce skeletal muscle hypertrophy in vivo

Supplementary Materials for

Crosstalk between Adiponectin and IGF-IR in breast cancer. Prof. Young Jin Suh Department of Surgery The Catholic University of Korea

Aberrant Promoter CpG Methylation is a Mechanism for Lack of Hypoxic Induction of

Supplementary Information. Glycogen shortage during fasting triggers liver-brain-adipose. neurocircuitry to facilitate fat utilization

The ubiquitin proteasome system in cardiovascular disease Basic mechanisms

Supplementary Table; Supplementary Figures and legends S1-S21; Supplementary Materials and Methods

RAS Genes. The ras superfamily of genes encodes small GTP binding proteins that are responsible for the regulation of many cellular processes.

Supplementary Figure 1.TRIM33 binds β-catenin in the nucleus. a & b, Co-IP of endogenous TRIM33 with β-catenin in HT-29 cells (a) and HEK 293T cells

Supplementary Figure 1. Baf60c and baf180 are induced during cardiac regeneration in zebrafish. RNA in situ hybridization was performed on paraffin

Supplementary Material

GW(g)/BW(g) GW(g)/BW(g) Con Dex Con Dex. GW(g)/BW(g) Relative mrna levels. Atrogin-1 Murf-1. Atrogin-1 Murf-1. Soleus

Protein phosphatase 2C-alpha knockdown reduces angiotensin II-mediated skeletal muscle wasting via restoration of mitochondrial recycling and function

Supplemental material for Hernandez et al. Dicoumarol downregulates human PTTG1/Securin mrna expression. through inhibition of Hsp90

T H E J O U R N A L O F C E L L B I O L O G Y

Validation & Assay Performance Summary

SUPPLEMENTARY INFORMATION

Legends for Supplemental Figures.

Supplemental Information

Data Sheet. Notch Pathway Reporter Kit Catalog # 60509

Construction of a hepatocellular carcinoma cell line that stably expresses stathmin with a Ser25 phosphorylation site mutation

Problem Set 8 Key 1 of 8

Molecular Medicine. Angiotensin II Induces Skeletal Muscle Atrophy by Activating TFEB-Mediated MuRF1 Expression

GH-Releasing Hormone Promotes Survival and Prevents TNF- -Induced Apoptosis and Atrophy in C2C12 Myotubes

Islet viability assay and Glucose Stimulated Insulin Secretion assay RT-PCR and Western Blot

Running Head: EFFECTS OF RESVERATROL ON CANCER CACHEXIA 1. Effects of resveratrol on cancer cachexia in a mouse model.

HSP72 HSP90. Quadriceps Muscle. MEF2c MyoD1 MyoG Myf5 Hsf1 Hsp GLUT4/GAPDH (AU)

supplementary information

Nature Medicine: doi: /nm.4324

/05/$15.00/0 Endocrinology 146(7): Copyright 2005 by The Endocrine Society doi: /en

Supplementary Figure 1 IMQ-Induced Mouse Model of Psoriasis. IMQ cream was

BIO360 Fall 2013 Quiz 1

Electrical Stimulation Control Nerve Regeneration via the p38 Mitogen-activated Protein Kinase and CREB

Not IN Our Genes - A Different Kind of Inheritance.! Christopher Phiel, Ph.D. University of Colorado Denver Mini-STEM School February 4, 2014

E3 ligase TRIM72 negatively regulates myogenesis by IRS-1 ubiquitination

J Jpn Coll Angiol, 2009, 49:

Supporting Information

Role of fatty acids in the development of insulin resistance and type 2 diabetes mellitus

Many catabolic conditions including diabetes

SUPPLEMENTARY INFORMATION

Plasma exposure levels from individual mice 4 hours post IP administration at the

The TWEAK-Fn14 System: Breaking the Silence of Cytokine- Induced Skeletal Muscle Wasting

Genetic Strain-Dependent Protein Metabolism and Muscle Hypertrophy Under Chronic Isometric Training in Rat Gastrocnemius Muscle

(A) PCR primers (arrows) designed to distinguish wild type (P1+P2), targeted (P1+P2) and excised (P1+P3)14-

Supplementary Figure 1. Normal T lymphocyte populations in Dapk -/- mice. (a) Normal thymic development in Dapk -/- mice. Thymocytes from WT and Dapk

Apoptosis is a cell-death program that plays an important

EFFECT OF ENDURANCE EXERCISE ALONE AND IN COMBINATION WITH IGF-1 ADMINISTRATION ON CELLULAR MARKERS INVOLVED IN SARCOPENIA.

FOR REVIEW. BMB Reports - Manuscript Submission. Manuscript Draft. Manuscript Number: BMB

Supplementary Materials and Methods

Supplementary Figure 1:

Supplementary Appendix

SUPPLEMENTARY INFORMATION

Supplementary Figure 1

Supplementary Figure 1. Establishment of prostacyclin-secreting hmscs. (a) PCR showed the integration of the COX-1-10aa-PGIS transgene into the

Signaling in the Nitrogen Assimilation Pathway of Arabidopsis Thaliana

Effects of Insulin-Like Growth Factor 1 on Muscle Atrophy and Motor Function in Rats with Brain Ischemia

CHAPTER 4 RESULTS. showed that all three replicates had similar growth trends (Figure 4.1) (p<0.05; p=0.0000)

SCIRF Award #2016 I-03 PI: Azizul Haque, PhD Grant Title: Neuron-specific Enolase and SCI

Reconciling data from transgenic mice that overexpress IGF-I specifically in skeletal muscle

Materials and Methods , The two-hybrid principle.

This student paper was written as an assignment in the graduate course

GLUCOCORTICOID-INDUCED MYOPATHY IS MEDIATED BY IMPAIRED NITRIC OXIDE SYNTHESIS

Supplementary information

Defective Hepatic Autophagy in Obesity Promotes ER Stress and Causes Insulin Resistance

Online Data Supplement. Anti-aging Gene Klotho Enhances Glucose-induced Insulin Secretion by Upregulating Plasma Membrane Retention of TRPV2

Transcription:

Am J Physiol Heart Circ Physiol 298: H1565 H1570, 2010. First published March 12, 2010; doi:10.1152/ajpheart.00146.2010. IGF-1 prevents ANG II-induced skeletal muscle atrophy via Akt- and Foxo-dependent inhibition of the ubiquitin ligase atrogin-1 expression Tadashi Yoshida, Laura Semprun-Prieto, Sergiy Sukhanov, and Patrice Delafontaine Heart and Vascular Institute, Tulane University School of Medicine, New Orleans, Louisiana Submitted 12 February 2010; accepted in final form 11 March 2010 Yoshida T, Semprun-Prieto L, Sukhanov S, Delafontaine P. IGF-1 prevents ANG II-induced skeletal muscle atrophy via Akt- and Foxo-dependent inhibition of the ubiquitin ligase atrogin-1 expression. Am J Physiol Heart Circ Physiol 298: H1565 H1570, 2010. First published March 12, 2010; doi:10.1152/ajpheart.00146.2010. Congestive heart failure is associated with activation of the reninangiotensin system and skeletal muscle wasting. Angiotensin II (ANG II) has been shown to increase muscle proteolysis and decrease circulating and skeletal muscle IGF-1. We have shown previously that skeletal muscle-specific overexpression of IGF-1 prevents proteolysis and apoptosis induced by ANG II. These findings indicated that downregulation of IGF-1 signaling in skeletal muscle played an important role in the wasting effect of ANG II. However, the signaling pathways and mechanisms whereby IGF-1 prevents ANG II-induced skeletal muscle atrophy are unknown. Here we show ANG II-induced transcriptional regulation of two ubiquitin ligases atrogin-1 and muscle ring finger-1 (MuRF-1) that precedes the reduction of skeletal muscle IGF-1 expression, suggesting that activation of atrogin-1 and MuRF-1 is an initial mechanism leading to skeletal muscle atrophy in response to ANG II. IGF-1 overexpression in skeletal muscle prevented ANG II-induced skeletal muscle wasting and the expression of atrogin-1, but not MuRF-1. Dominant-negative Akt and constitutively active Foxo-1 blocked the ability of IGF-1 to prevent ANG IImediated upregulation of atrogin-1 and skeletal muscle wasting. Our findings demonstrate that the ability of IGF-1 to prevent ANG IIinduced skeletal muscle wasting is mediated via an Akt- and Foxo- 1-dependent signaling pathway that results in inhibition of atrogin-1 but not MuRF-1 expression. These data suggest strongly that atrogin-1 plays a critical role in mechanisms of ANG II-induced wasting in vivo. angiotensin II; insulin-like growth factor-1 Address for reprint requests and other correspondence: P. Delafontaine, Heart and Vascular Institute, Tulane Univ. School of Medicine, 1430 Tulane Ave. SL-48, New Orleans, LA 70112 (e-mail: pdelafon@tulane.edu). CONGESTIVE HEART FAILURE (CHF) is a leading cause of cardiovascular mortality and morbidity (8). CHF is associated with elevated circulating levels of angiotensin II (ANG II) (10) and muscle wasting, which is an important predictor of poor outcome in patients with this disease (2, 22). We have demonstrated that ANG II infusion in mice stimulates muscle protein wasting by activating proteolytic pathways involving caspase-3 and the ubiquitin-proteasome system, accompanied by depression of circulating and skeletal muscle insulin-like growth factor-1 (IGF-1). These findings suggested that downregulation of IGF-1 signaling in skeletal muscle played an important role in the wasting effect of ANG II. Studies using in vitro models of muscle atrophy such as dexamethasone-induced proteolysis have indicated that IGF-1 acts through Akt and Foxo transcription factors to suppress expression of the two E3 ubiquitin ligase genes atrogin-1 and muscle ring finger-1 (MuRF-1) (16). Atrogin-1 and MuRF-1 are highly induced in various muscle atrophy models (7). We have shown that in transgenic mice overexpressing IGF-1 specifically in skeletal muscle ANG II-induced caspase-3 activation, protein ubiquitination and muscle atrophy are inhibited (20). In the present study we used electroporation of plasmid constructs in vivo to determine signaling mechanisms whereby IGF-1 inhibits ANG II-induced atrophy. Our data demonstrate that IGF-1 prevents ANG II-induced skeletal muscle wasting through an Akt/Foxo-dependent pathway in vivo and suggest an important role for atrogin-1 in ANG II effects on skeletal muscle. MATERIALS AND METHODS Materials. Recombinant human IGF-1 was from Tercica. Antibodies against phospho-akt (Ser473), phospho-foxo-1 (Ser256), total Akt, and total Foxo-1 were purchased from Cell Signaling Technology. Human IGF-1 expression plasmid is a generous gift from Dr. Thissen (17). dnakt and cafoxo-1 expression plasmids were constructed from adenoviral expression vectors described previously (7, 19). Animal studies. Animal protocols were approved by the Institutional Animal Care and Use Committee at Tulane University. Nine- to ten-week-old male FVB (Charles River), C57BL/6 (Charles River), and IGF-1-transgenic [myosin light chain (MLC)/mIgf-1] mice were used in this study. Osmotic minipumps (Alzet model 1007D; Durect) were implanted to infuse ANG II at a rate of 1 mg kg 1 min 1 for FVB and MLC/mIgf-1 and 1.5 mg kg 1 min 1 for C57BL/6, respectively. The body weight and food intake of each ANG II-infused mouse were measured daily, and a corresponding vehicle-infused control mouse was given the same amount of food as that eaten by the ANG II-infused mouse on the previous day (i.e., mice were pair fed). Blood pressures were measured noninvasively using the Visitech BP-2000 (Visitech Systems) as previously described (4). One or seven days after implantation, mice were anesthetized and tissues were removed, weighed, and snap-frozen in liquid nitrogen and stored at 80 C until processed. Skeletal muscle electroporation. Two weeks before ANG II minipump implantation, each mouse was anesthetized with a mixture of 75 mg/kg ketamine (Ketalar; Pfizer, Oslo, Norway) and 15 mg/kg xylazine hydrochloride (Rompun; Bayer, Fernwald, Germany) administered by injection intraperitoneally. Mice hindlimbs were shaved, and plasmids (1 g/ l, dissolved in saline) were injected into five different sites (total volume per muscle 50 l) in each gastrocnemius muscle with a 22-gauge needle syringe (Hamilton model 705). Transcutaneous pulses were applied by two stainless steel plate electrodes (Caliper Electrode model 384; BTX) with a distance between the two plates of 0.5 cm, and conductive gel was used to ensure the electrical contact. Electric pulses with a standard square wave were delivered by an electroporator (ECM830 Electro Square Porator; BTX). Eight pulses of 100 V/cm were administered to the muscle with a delivery rate of 1 pulse/s with each pulse 20 ms in duration. Quantitative real-time RT-PCR. Gastrocnemius muscles were homogenized in Tripure isolation reagent (Roche), and total RNA was isolated following manufacturer s instructions. cdna synthesis was performed using the RT2 first-strand kit (SABiosciences, Frederick, MD). Real-time PCR was performed using a 40-cycle two-step PCR http://www.ajpheart.org 0363-6135/10 $8.00 Copyright 2010 the American Physiological Society H1565

H1566 protocol in the icycler IQ real-time detection system (Bio-Rad, Hercules, CA). Hprt1 gene expression was used as an internal control (14). PCR primers used in this study were obtained commercially [SABiosciences; PPM38061A for atrogin-1, PPM61045A for MuRF-1, PPM03387E for IGF-1, PPM04714E for IGF-1 receptor (IGF-1R), and PPM03559E for Hprt1]. Immunoblotting. Samples were solubilized in radioimmunoprecipitation assay buffer, and proteins were separated by electrophoresis on 10% SDS-polyacrylamide gels. Proteins were blotted onto polyvinylidene difluoride membranes and, after being blocked with 5% BSA/ 0.1% Tween 20, incubated with primary antibodies in the same solution. Bound antibodies were detected by anti-rabbit or -mouse IgG conjugated with peroxidase and subsequently detected by chemiluminescence. Luciferase reporter gene assay. 5 kbp and 1 kbp upstream promoter regions of atrogin-1 and MuRF-1 were cloned from C57BL/6 mouse genomic DNA by PCR using the following primers: 5=-GGCGGTACCTGCGGCATCCATAGCTCTGTC-3= and 5=-GCG- GTCGACGGTACAGAGCGCGGACGCGAC-3= for atrogin-1 5kbp; 5=-GGCGGTACCCGCCTCGACGCCAGCGCTGC-3= and 5=- GCGGTCGACGGTACAGAGCGCGGACGCGAC-3= for atrogin-1 1kbp; 5=-GCGGTCGACCTCCAGAGCTGTCAGGCTATC-3= and 5=-GCGCTCGAGATTGCTTTGAAATGCGACTGTC-3= for MuRF-1 5kbp; 5=-GCGGTCGACGGAATATAGACTTGCAGATC-3= and 5=- GCGCTCGAGATTGCTTTGAAATGCGACTGTC-3= for MuRF-1 1kbp. These DNA fragments were subcloned into pgl4.12[luc2cp] (Promega). Twenty micrograms of plasmids containing atrogin-1 or MuRF-1 promoter sequences were electroporated into gastrocnemius muscle together with 2 g of pgl4.74[hrluc/tk] vector (Promega), followed by ANG II minipump implantation 2 wk after electroporation. Gastrocnemius muscles were collected at day 1 of the infusion. Skeletal muscle lysates were prepared, and luciferase activity was measured by Dual-Luciferase Reporter Assay System (Promega) following the manufacturer s instruction. Statistical analysis. All data represent means SE of at least five animals in each group, and results were analyzed using Student s t-test when data from two experimental groups were compared or ANOVA followed by Bonferroni s multiple comparison test when data from three groups were studied. RESULTS ANG II-induced upregulation of ubiquitin ligase expression precedes IGF-1 downregulation; differential regulation of ubiquitin ligases in MLC/mIgf-1 mice. We previously reported that after 7 days of ANG II infusion in mice, IGF-1 mrna levels were reduced but mrna levels of the ubiquitin ligases atrogin-1 and MuRF-1 were increased in skeletal muscle (20, 23). Since IGF-1 has been reported to suppress expression of atrogin-1 and MuRF-1 (21), our findings suggested that downregulation of IGF-1 could be involved in upregulation of ubiquitin ligase expression. We determined the time coursedependent expression of these genes in the skeletal muscle of ANG II-infused mice using quantitative PCR. Atrogin-1 and MuRF-1 expression was rapidly upregulated within 1 day after ANG II infusion, followed by a gradual reduction over 7 days (Fig. 1, A and B). On the other hand, reduced IGF-1 expression was only seen 7 days after infusion (Fig. 1C), indicating that upregulation of ubiquitin ligase expression was unlikely to result from reduced IGF-1 signaling. IGF-1R expression increased 1 and 4 days after infusion and returned to basal levels after 7 days (Fig. 1D). MLC/mIgf-1 mice overexpress a skeletal muscle-specific isoform of IGF-1 preferentially in fast fiber muscles under the control of the myosin light chain promoter (13) and are resistant to ANG II-induced skeletal Fig. 1. Regulation of gene expression in skeletal muscle of ANG II-infused wild-type and myosin light chain (MLC)/mIgf-1 mice. ANG II or saline minipumps were implanted to FVB or MLC/mIgf-1 mice, and gastrocnemius muscles were collected 1, 4, and 7 days after implantation. Gastrocnemius muscle weight of each animal was measured 7 days after minipump implantation (E), and atrogin-1 (A), muscle ring finger-1 (MuRF-1; B), IGF-1 (C), and IGF-1 receptor (IGF-1R; D) expression was quantified by quantitative RT- PCR on days 1, 4, and 7 of infusion. Gastrocnemius muscles of FVB and MLC/mIgf-1 mice were collected 1 day after ANG II or saline infusion, and atrogin-1 (F) and MuRF-1 (G) expression was measured by quantitative RT-PCR. Means are SE; n 5to6.*P 0.05; **P 0.01. muscle atrophy (20). In gastrocnemius muscle of MLC/mIgf-1 mice, basal expression of atrogin-1 was significantly repressed, and ANG II infusion failed to increase its expression (Fig. 1F). On the other hand, neither basal nor ANG II-induced MuRF-1 expression was affected in skeletal muscle of MLC/mIgf-1 mice (Fig. 1G). Consistent with our previous data, the ANG II-induced reduction in gastrocnemius weight was inhibited in MLC/mIgf-1 mice (Fig. 1E). IGF-1 plasmid electroporation blocks atrogin-1 but not MuRF-1 upregulation in ANG II-infused mice. Electroporation of plasmid constructs has been reported to be a useful technol-

ogy for the analysis of signaling pathways involved in skeletal muscle hypertrophy and atrophy (17, 18). We determined the efficiency of gene expression by using enhanced green fluorescent protein-encoding plasmid electroporated to gastrocnemius muscle and observed highly efficient and long-lasting expression as reported previously (Fig. 2, A and B) (1, 12, 17, 18). However, we found that the electric pulse associated with plasmid electroporation to gastrocnemius muscle significantly increased atrogin-1 and MuRF-1 mrna levels, whereas plasmid injection alone had no effect on gene expression (Fig. 2, E and F). Furthermore, electroporation was associated with a marked but transient increase in -actin expression (Fig. 2B). The increase of ubiquitin ligase gene expression reached the maximal level 1 day after electroporation, followed by a gradual decrease and return to basal levels 1 to 2 wk after electroporation (Fig. 2, G and H). These data indicate that a recovery period is required after plasmid electroporation to analyze the signaling pathways associated with skeletal muscle atrophy. Since the elevation of atrogin-1 and MuRF-1 completely returned to basal levels after 2 wk of electroporation, we chose a 2-wk recovery period after plasmid electroporation before minipump implantation. Human IGF-1 (higf-1) mrna levels were highly upregulated in higf-1 plasmid electroporated gastrocnemius muscles (Fig. 2C), without significant detection of plasma higf-1 (Fig. 2D). Following electroporation of higf-1 or control empty vector into mouse gastrocnemius muscle and the 2-wk recovery period, mice were implanted with ANG II or saline minipumps and skeletal muscle weight and ubiquitin ligase expression were measured 1 and 7 days after infusion. As reported previously, ANG II infusion caused a loss of body weight in pair-fed mice (P 0.01 by ANOVA compared with sham; Fig. 3A) and reduced food intake (Fig. 3B). The body weight change in the sham group before and after minipump implantation was not significant. Local higf-1 expression prevented the loss of skeletal muscle weight induced by ANG II (Fig. 3C) and markedly suppressed ANG II-induced increase of atrogin-1 expression (Fig. 3D). On the other hand, MuRF-1 expression caused by ANG II was not affected by local higf-1 overexpression (Fig. 3E). To confirm these findings and to obtain initial insights into mechanisms whereby ANG II increases ubiquitin ligase mrna levels, we generated luciferase reporter gene constructs that contain 5 kbp and 1 kbp of atrogin-1 and MuRF-1 upstream promoter regions and analyzed these promoter activities in gastrocnemius muscle (Fig. 3F). Our data showed that ANG II induction of atrogin-1 and MuRF-1 transcriptional activity was mediated by 1 kbp and 5 kbp 5= upstream regions of atrogin-1 and MuRF-1, respectively. Electroporation of IGF-1 expression plasmids together with these luciferase vectors showed that IGF-1 suppressed ANG II-induced atrogin-1 promoter activity, and this was the case using both 5 kbp and 1 kbp constructs. However, IGF-1 coexpression failed to suppress ANG II-induced MuRF-1 promoter activity. IGF-1 prevents ANG II-induced skeletal muscle atrophy via Akt-Foxo signaling. It has been previously demonstrated that induction of myotube hypertrophy by IGF-1 depends on phosphatidylinositol 3-kinase-mediated activation of Akt (15). Akt phosphorylates the Forkhead box (Foxo) class of transcription factors, resulting in their nuclear exclusion, thereby inhibiting their transcriptional activity. Activation of the IGF-1-PI3K- H1567 Fig. 2. Efficiency and deleterious effect of plasmid electroporation into mouse gastrocnemius muscle. A: enhanced green fluorescent protein (EGFP) fluorescence after electroporation. EGFP encoding plasmid was electroporated to gastrocnemius muscle, and fluorescence was observed 3, 5, 7, and 14 days after electroporation. Whole gastrocnemius muscle is shown in the picture. Contralateral control gastrocnemius muscle was electroporated with empty vector. B: EGFP expression in gastrocnemius muscle after EGFP encoding plasmid electroporation was assessed by immunoblotting. C: IGF-1 mrna expression was analyzed by Northern blotting 5 days after electroporation. Endogenous and plasmid-derived IGF-1 transcripts are indicated by white and black arrow, respectively. I, IGF-1 plasmid; C, control plasmid. D: plasma human IGF-1 (higf-1) levels were quantified before and after electroporation (EP) of IGF-1 encoding plasmid. Plasma samples collected from mice infused (inf) with 1.5 mg kg 1 day 1 human IGF-1 were used as control. E and F: atrogin-1 and MuRF-1 expression was quantified 1 day after electroporation by quantitative RT-PCR. Gastrocnemius muscles were collected from empty plasmid vector electroporated (inj EP), plasmid vector injected without electric pulse (inj), or electric pulse applied without plasmid injection (EP) skeletal muscles. Neither plasmid injection nor electric pulse administration was performed in control muscles. G and H: atrogin-1 and MuRF-1 expression were quantified 1, 3, 7, and 14 days after electroporation. Empty plasmid vector was injected in both of the muscles, and electric pulse was applied only to the EP gastrocnemius muscles. Means are SE; n 5. *P 0.05; **P 0.01.

H1568 Akt-Foxo pathway inhibits skeletal muscle atrophy induced by starvation, denervation, and glucocorticoids (16 18, 21). To determine the downstream signaling mechanisms mediating IGF-1 prevention of ANG II-induced skeletal muscle atrophy, we used dominant-negative and constitutively active Akt and Foxo-1 variants. The dominant-negative form of Akt (dnakt) has alternative residues substituted for lysine at position 179, threonine at position 308, and serine at position 473 (7, 19) and when electroporated to gastrocnemius muscle markedly reduced levels of phosphorylated Akt (Fig. 4A). Electroporation of dnakt, but not of wild-type Akt, along with IGF-1 blocked the ability of IGF-1 to prevent ANG II-induced loss of skeletal muscle weight (Fig. 4B). Furthermore, IGF-1 failed to inhibit ANG II-induced atrogin-1 gene expression in the presence of dnakt (Fig. 4C). On the other hand, ANG II-induced MuRF-1 expression was not affected by dnakt expression (Fig. 4D). The constitutively active form of Foxo-1 (cafoxo-1) has alanine residues substituted for those serine/threonine residues that are phosphorylated by Akt, which prevents its inactivation by phosphorylation. Similarly to the results obtained using dnakt, when cafoxo-1 was electroporated to gastrocnemius muscle, IGF-1 could not counteract the ANG II-induced loss of skeletal muscle weight (Fig. 5B). Furthermore, cafoxo-1 upregulated basal levels of atrogin-1 mrna in sham-infused animals and prevented the IGF-1-mediated repression of atrogin-1 gene expression in ANG II-infused mouse skeletal muscle (Fig. 5C). However, neither basal nor ANG II-induced MuRF-1 expression was affected by cafoxo-1 expression (Fig. 5D). DISCUSSION We have previously shown that ANG II induces weight loss in part via a catabolic effect on skeletal muscle that is characterized by downregulation of IGF-1 expression, reduced pakt and pfoxo expression, and upregulation of ubiquitin ligase expression (20). In MLC/mIgf-1 mice the ANG II induction of weight loss was blocked, and this effect was accompanied by maintenance of pakt levels, suggesting potential involvement of the Akt-Foxo pathway in the ability of IGF-1 to prevent Fig. 3. Electroporation of IGF-1 to skeletal muscle inhibits ANG II-induced skeletal muscle wasting and atrogin-1 but not MuRF-1 expression. C57BL/6 mice were electroporated with plasmid encoding IGF-1 or control empty vector into each gastrocnemius muscle, followed by 2-wk recovery period (day 14 to day 0). ANG II or saline infusion was started on day 0, and body weight (A) and food intake (B) were measured daily. Saline infused mice were pair fed. Gastrocnemius muscle weight of plasmid electroporated and ANG II or sham infused mice were measured on day 7 of infusion (C). Atrogin-1 (D) and MuRF-1 (E) expression in plasmid electroporated gastrocnemius muscle was quantified by quantitative RT-PCR after 1 day of ANG II infusion. F: activity of 5 and 1 kbp upstream promoter regions of atrogin-1 and MuRF-1 were measured by dual luciferase reporter gene assay. Firefly luciferase activity of gastrocnemius muscle collected 1 day after ANG II minipump implantation was determined, and Renilla luciferase under the control of thymidine kinase was used as an internal electroporation control. Luciferase activity was calculated as the ratio of firefly and Renilla luciferase bioluminescence. Means are SE; n 5. *P 0.05; **P 0.01. AU, arbitrary units. Fig. 4. IGF-1 inhibition of ANG II muscle wasting and atrogin-1 expression is blocked by dominant-negative form of Akt (dnakt). A: plasmids encoding wild-type Akt (wtakt) or dnakt were electroporated to gastrocnemius muscle, and phosphorylated and total Akt levels were analyzed by immunoblotting 7 days after electroporation. B D: plasmid encoding IGF-1, together with wtakt or dnakt encoding vectors, was electroporated to gastrocnemius muscle, and ANG II or saline minipumps were implanted 2 wk after electroporation. Gastrocnemius muscles were collected 1 or 7 days after infusion. The weight of each plasmid electroporated gastrocnemius muscle (B) was measured 1 wk after minipump implantation. Atrogin-1 (C) and MuRF-1 (D) expression in gastrocnemius muscle was quantified by quantitative RT-PCR 1 day after minipump implantation. Means are SE; n 5. *P 0.05; **P 0.01.

Fig. 5. IGF-1 inhibition of ANG II muscle wasting and atrogin-1 expression is blocked by constitutively active form of Foxo-1 (cafoxo-1). A: plasmids encoding wild-type (wt)foxo-1 or cafoxo-1 were electroporated to gastrocnemius muscle, and phosphorylated and total Foxo-1 levels were analyzed by immunoblotting 7 days after electroporation. B D: plasmid encoding IGF-1, together with wtfoxo-1 or cafoxo-1 encoding vectors, was electroporated to gastrocnemius muscle, and ANG II or saline minipumps were implanted 2 wk after electroporation. Gastrocnemius muscles were collected 1 or 7 days after infusion. Weight of each plasmid electroporated gastrocnemius muscle (B) was measured 1 wk after minipump implantation. Atrogin-1 (C) and MuRF-1 (D) expression in gastrocnemius muscle was quantified by quantitative RT-PCR 1 day after minipump implantation. Means are SE; n 5. *P 0.05; **P 0.01. ANG II-induced atrophy. Our current study establishes that Akt-Foxo-dependent signaling is required for the rescue effect of IGF-1. Furthermore, the rapid induction of ubiquitin ligase gene expression by ANG II and the ability of IGF-1 electroporation to block ANG II upregulation of atrogin-1 expression suggest strongly that atrogin-1 plays a critical role in the catabolic effect of ANG II. Because IGF-1 has been shown to repress atrogin-1 expression in vivo in other catabolic states (13, 21) we hypothesized that the reduction in IGF-1 expression induced by ANG II could have resulted in upregulation of ubiquitin ligase expression. However, time course analysis (Fig. 1) clearly showed that the rapid upregulation of atrogin-1 and MuRF-1 in response to ANG II occurred within 24 h and preceded downregulation of IGF-1. Thus upregulation of ubiquitin ligase expression is likely to play an important role in early mechanisms leading to loss of muscle in response to ANG II and is followed by reduced IGF-1 expression and signaling, which likely contributes to ongoing loss of muscle. To determine the role of the Akt and Foxo pathways in the ability of IGF-1 to prevent ANG II muscle loss, we electroporated IGF-1 plasmid together with dnakt and cafoxo-1 plasmids. The blocking effect of dnakt on IGF-1 inhibition of ANG II-induced atrogin-1 expression and muscle wasting (Fig. 4) demonstrates that Akt is required for the rescue effect of IGF-1. Akt is a serine/threonine kinase with multiple downstream targets including Foxo, BCL2-antagonist of death, caspase-9, glycogen synthase kinase-3, and tuberous sclerosis 2 (6). The Foxo family consists of four members in mammalian H1569 cells: Foxo-1, Foxo-3a, Foxo-4, and Foxo-6. Foxo-1 and Foxo-3 have received the most attention with regard to skeletal muscle atrophy because they are upregulated in various models of atrophy (9). The ability of cafoxo to inhibit the rescue effect of IGF-1 (Fig. 5) is consistent with a model in which IGF-1- induced phosphorylation and inactivation of Foxo-1 protein via an Akt signaling pathway plays a critical role in the ability of IGF-1 to inhibit ANG II-induced ubiquitin ligase expression and muscle wasting. IGF-1 inhibition of muscle atrophy in other catabolic conditions (5, 13, 17) has been suggested to be due to its ability to repress expression of atrogin-1 and MuRF-1. Our findings using MLC/mIgf-1 mice and electroporation of IGF-1 plasmid clearly showed that overexpression of IGF-1 in skeletal muscle blocks ANG II upregulation of atrogin-1, but not MuRF-1. This finding is surprising since in most other catabolic conditions these ubiquitin ligases are coordinately regulated (3, 11). For instance, in dexamethasone-induced atrophy intramuscular injection of IGF-1 prevented Akt/Foxo signaling, resulting in repression of atrogin-1 and MuRF-1 expression (17). However, it is important to note that in NF- B-mediated muscle atrophy there is an increase in MuRF-1 expression but no change in atrogin-1 (5). To confirm our findings and to obtain initial insights into mechanisms whereby ANG II upregulates ubiquitin ligase expression, we cloned upstream promoter sequences from both atrogin-1 and MuRF-1 mouse genomic DNA and generated promoter reporter gene constructs. Our data clearly demonstrate marked upregulation of reporter gene expression in the skeletal muscles of ANG II-infused animals and abrogation of atrogin-1 but not MuRF-1 luciferase activity following electroporation of IGF-1. These finding unequivocally demonstrate that ANG II transcriptionally regulates atrogin-1 /MuRF-1 expression but that the rescue effect of IGF-1 is characterized by inhibition of atrogin-1 but not MuRF-1, strongly suggesting specific involvement of atrogin-1 but not MuRF-1 in ANG II-induced wasting. This is schematically represented in Fig. 6. To our knowledge this is the first report of a catabolic condition in which IGF-1 differentially represses atrogin-1 Fig. 6. Signaling pathways involved in ANG II-induced skeletal muscle atrophy and its prevention by IGF-1. ANG II has been shown to cause dephosphorylation of Akt. Activation of Foxo results in transcriptional upregulation of atrogin-1. IGF-1 activates Akt resulting in phosphorylation and inactivation of Foxo and inhibition of atrogin-1 transcription. Although MuRF-1 is also activated by ANG II in skeletal muscle, its transcription is not affected by IGF-1 or Akt-Foxo signaling.

H1570 expression without altering MuRF-1 expression. It is of note that little is known about transcriptional control of atrogin-1 and MuRF-1 expression, although Foxo-3 has been shown to stimulate transcription of the atrogin-1 promoter (16). Our findings demonstrate that ANG II infusion in mice strongly increases skeletal muscle atrogin-1 and MuRF-1 mrna levels, likely in large part via a transcriptional effect. However, this effect of ANG II is unlikely to be a direct effect since we have found that adult rodent skeletal muscles express little to no ANG II receptors (23). Thus it is more likely that the ANG II upregulation of ubiquitin ligase expression is mediated via other molecules such as glucocorticoids, IL-6, and serum amyloid A that we have shown to be involved in mediating the catabolic effects of ANG II (20, 23). A novel aspect of our study is the discovery that the electric pulse application associated with plasmid electroporation into skeletal muscle is sufficient to increase ubiquitin ligase expression for up to 1 wk (Fig. 2, G and H). This effect likely results from an injury response since -actin expression was also transiently increased in electroporated muscle (Fig. 2B). Because electroporation is increasingly applied for the study of skeletal muscle signaling pathways (1, 13, 17, 18), our findings have important implications for the application of this technology and indicate that an appropriate recovery period is critical, particularly for the study of skeletal muscle atrophy-associated signaling. In summary, we have shown that the upregulation of atrogin-1 and MuRF-1 expression in skeletal muscles of ANG II-infused mice is mediated via a transcriptional effect that precedes ANG II-induced downregulation of IGF-1 expression. The ability of IGF-1 to prevent ANG II-induced skeletal muscle wasting is mediated via an Akt- and Foxo-1-dependent signaling pathway that results in inhibition of atrogin-1 but not MuRF-1 expression. These data suggest strongly that atrogin-1 plays a critical role in mechanisms of ANG II-induced wasting in vivo. ACKNOWLEDGMENTS We acknowledge Dr. Nadia Rosenthal for providing MLC/mIgf-1 mice and Dr. Jean-Paul Thissen for human IGF-1 expression plasmid. GRANTS This work was supported by National Heart, Lung, and Blood Institute Grants R01-HL-070241 (to P. Delafontaine) and R01-HL-080682 (to P. Delafontaine) and the Tulane Research Enhancement Fund (to P. Delafontaine). DISCLOSURES No conflicts of interest, financial or otherwise, are declared by the author(s). REFERENCES 1. Aihara H, Miyazaki J. Gene transfer into muscle by electroporation in vivo. Nat Biotechnol 16: 867 870, 1998. 2. Anker SD, Ponikowski P, Varney S, Chua TP, Clark AL, Webb- Peploe KM, Harrington D, Kox WJ, Poole-Wilson PA, Coats AJ. Wasting as independent risk factor for mortality in chronic heart failure. Lancet 349: 1050 1053, 1997. 3. Attaix D, Ventadour S, Codran A, Bechet D, Taillandier D, Combaret L. The ubiquitin-proteasome system and skeletal muscle wasting. Essays Biochem 41: 173 186, 2005. 4. Brink M, Wellen J, Delafontaine P. Angiotensin II causes weight loss and decreases circulating insulin-like growth factor I in rats through a pressor-independent mechanism. J Clin Invest 97: 2509 2516, 1996. 5. Cai D, Frantz JD, Tawa NE Jr, Melendez PA, Oh BC, Lidov HG, Hasselgren PO, Frontera WR, Lee J, Glass DJ, Shoelson SE. IKKbeta/ NF-kappaB activation causes severe muscle wasting in mice. Cell 119: 285 298, 2004. 6. Franke TF. PI3K/Akt: getting it right matters. Oncogene 27: 6473 6488, 2008. 7. Fujio Y, Guo K, Mano T, Mitsuuchi Y, Testa JR, Walsh K. Cell cycle withdrawal promotes myogenic induction of Akt, a positive modulator of myocyte survival. Mol Cell Biol 19: 5073 5082, 1999. 8. Iaina A, Silverberg DS, Wexler D. Therapy insight: congestive heart failure, chronic kidney disease and anemia, the cardio-renal-anemia syndrome. Nat Clin Pract Cardiovasc Med 2: 95 100, 2005. 9. Kandarian SC, Jackman RW. Intracellular signaling during skeletal muscle atrophy. Muscle Nerve 33: 155 165, 2006. 10. Kuroda T, Shida H. Angiotensin II-induced myocardial damage with a special reference to low cardiac output syndrome. Jpn Heart J 24: 235 243, 1983. 11. Lecker SH. Ubiquitin-protein ligases in muscle wasting: multiple parallel pathways? Curr Opin Clin Nutr Metab Care 6: 271 275, 2003. 12. Mir LM, Bureau MF, Gehl J, Rangara R, Rouy D, Caillaud JM, Delaere P, Branellec D, Schwartz B, Scherman D. High-efficiency gene transfer into skeletal muscle mediated by electric pulses. Proc Natl Acad Sci USA 96: 4262 4267, 1999. 13. Musaro A, McCullagh K, Paul A, Houghton L, Dobrowolny G, Molinaro M, Barton ER, Sweeney HL, Rosenthal N. Localized Igf-1 transgene expression sustains hypertrophy and regeneration in senescent skeletal muscle. Nat Genet 27: 195 200, 2001. 14. Nishimura M, Nikawa T, Kawano Y, Nakayama M, Ikeda M. Effects of dimethyl sulfoxide and dexamethasone on mrna expression of housekeeping genes in cultures of C2C12 myotubes. Biochem Biophys Res Commun 367: 603 608, 2008. 15. Rommel C, Bodine SC, Clarke BA, Rossman R, Nunez L, Stitt TN, Yancopoulos GD, Glass DJ. Mediation of IGF-1-induced skeletal myotube hypertrophy by PI3K/Akt/mTOR and PI3K/Akt/GSK3 pathways. Nat Cell Biol 3: 1009 1013, 2001. 16. Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, Walsh K, Schiaffino S, Lecker SH, Goldberg AL. Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell 117: 399 412, 2004. 17. Schakman O, Gilson H, de Coninck V, Lause P, Verniers J, Havaux X, Ketelslegers JM, Thissen JP. Insulin-like growth factor-i gene transfer by electroporation prevents skeletal muscle atrophy in glucocorticoidtreated rats. Endocrinology 146: 1789 1797, 2005. 18. Schakman O, Kalista S, Bertrand L, Lause P, Verniers J, Ketelslegers JM, Thissen JP. Role of Akt/GSK-3beta/beta-catenin transduction pathway in the muscle anti-atrophy action of insulin-like growth factor-i in glucocorticoid-treated rats. Endocrinology 149: 3900 3908, 2008. 19. Skurk C, Maatz H, Kim HS, Yang J, Abid MR, Aird WC, Walsh K. The Akt-regulated forkhead transcription factor FOXO3a controls endothelial cell viability through modulation of the caspase-8 inhibitor FLIP. J Biol Chem 279: 1513 1525, 2004. 20. Song YH, Li Y, Du J, Mitch WE, Rosenthal N, Delafontaine P. Muscle-specific expression of IGF-1 blocks angiotensin II-induced skeletal muscle wasting. J Clin Invest 115: 451 458, 2005. 21. Stitt TN, Drujan D, Clarke BA, Panaro F, Timofeyva Y, Kline WO, Gonzalez M, Yancopoulos GD, Glass DJ. The IGF-1/PI3K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin ligases by inhibiting FOXO transcription factors. Mol Cell 14: 395 403, 2004. 22. Strassburg S, Springer J, Anker SD. Muscle wasting in cardiac cachexia. Int J Biochem Cell Biol 37: 1938 1947, 2005. 23. Zhang L, Du J, Hu Z, Han G, Delafontaine P, Garcia G, Mitch WE. IL-6 and serum amyloid A synergy mediates angiotensin II-induced muscle wasting. J Am Soc Nephrol 20: 604 612, 2009.