Research ArticleMuscle Mechanics and Ventricular Function

Desmin loss and mitochondrial damage precede left ventricular systolic failure in volume overload heart failure

Published Online:https://doi.org/10.1152/ajpheart.00027.2017

Abstract

Heart failure due to chronic volume overload (VO) in rats and humans is characterized by disorganization of the cardiomyocyte desmin/mitochondrial network. Here, we tested the hypothesis that desmin breakdown is an early and continuous process throughout VO. Male Sprague-Dawley rats had aortocaval fistula (ACF) or sham surgery and were examined 24 h and 4 and 12 wk later. Desmin/mitochondrial ultrastructure was examined by transmission electron microscopy (TEM) and immunohistochemistry (IHC). Protein and kinome analysis were performed in isolated cardiomyocytes, and desmin cleavage was assessed by mass spectrometry in left ventricular (LV) tissue. Echocardiography demonstrated a 40% decrease in the LV mass-to-volume ratio with spherical remodeling at 4 wk with ACF and LV systolic dysfunction at 12 wk. Starting at 24 h and continuing to 4 and 12 wk, with ACF there is TEM evidence of extensive mitochondrial clustering, IHC evidence of disorganization associated with desmin breakdown, and desmin protein cleavage verified by Western blot analysis and mass spectrometry. IHC results revealed that ACF cardiomyocytes at 4 and 12 wk had perinuclear translocation of αB-crystallin from the Z disk with increased α, β-unsaturated aldehyde 4-hydroxynonelal. Use of protein markers with verification by TUNEL staining and kinome analysis revealed an absence of cardiomyocyte apoptosis at 4 and 12 wk of ACF. Significant increases in protein indicators of mitophagy were countered by a sixfold increase in p62/sequestosome-1, which is indicative of an inability to complete autophagy. An early and continuous disruption of the desmin/mitochondrial architecture, accompanied by oxidative stress and inhibition of apoptosis and mitophagy, suggests its causal role in LV dilatation and systolic dysfunction in VO.

NEW & NOTEWORTHY This study provides new evidence of early onset (24 h) and continuous (4−12 wk) desmin misarrangement and disruption of the normal sarcomeric and mitochondrial architecture throughout the progression of volume overload heart failure, suggesting a causal link between desmin cleavage and mitochondrial disorganization and damage.

a primary volume overload (VO), as observed in primary mitral regurgitation (MR) or aortocaval fistula (ACF), induces left ventricular (LV) diastolic overload without an increase in systolic blood pressure (25). This pure stretch on the myocardium results in adverse eccentric LV remodeling characterized by a progressive LV spherical dilatation, LV wall thinning, and cardiomyocyte thinning with elongation (11, 45, 48). Initial studies have focused on the loss of the extracellular matrix in the pathophysiology of adverse eccentric LV remodeling in a primary VO (11, 45, 54, 57). Our recent studies have demonstrated that this extracellular matrix breakdown and loss is matched by cleavage and loss of organization of major cytoskeletal intermediate filament desmin, along with myofibrillar degeneration, mitochondrial clustering, disorganization, and cristae disruption, both in rats with chronic ACF (56) and in humans with chronic MR and preserved LV ejection fraction (LVEF) (4). We have further linked mitochondrial production of ROS with desmin breakdown in LV cardiomyocytes of people with MR (3, 4) and rats with ACF (22) and in isolated cardiomyocytes undergoing cyclical stretch (22, 56).

Currently, nearly 70 mutations have been identified in the desmin or αB-crystallin genes that alter the desmin filament assembly process (8, 10, 27, 34) and result in the disorganization of the desmin/mitochondrial network and accumulation of desmin-containing aggregates. However, there is now emerging evidence that excessive adrenergic drive and PKC activation are associated with phosphorylation of desmin and its subsequent cleavage in nongenetic causes of human heart failure in humans and in animal models (1, 28). Indeed, in addition to our recent report of patients with isolated MR, studies of patients with ischemic cardiomyopathy have highlighted the disruption and patchy loss of desmin in LV cardiomyocytes and linked them with LV dysfunction and poor prognosis (12, 13). In a canine model of dyssynchronous heart failure, Agnetti and coworkers (1) postulated that a decrease in cardiac output produces a deleterious increase in G protein-coupled receptor signaling that resulted in increased desmin cleavage, which was reversed by restoration of cardiac synchrony.

Another important consequence of a pure VO, in which the excess volume is ejected into the low-pressure atrium or arteriovenous fistula, is the early and incessant activation of the adrenergic nervous system, which has been documented in humans (24, 35, 58) and dogs (26, 38) with MR and in rats (53, 54) with ACF. In patients with chronic MR (4) and in rats with ACF at 8 wk (56), there is extensive desmin breakdown as revealed by immunohistochemistry and Western blot analysis and cytoskeletal/mitochondrial disruption as revealed by transmission electron microscopy (TEM). The similarities in human MR and rat ACF allow for clinically relevant animal studies that test unique pharmacologic interventions and provide a better understanding for the unexplained decrease in LV systolic function after mitral valve repair/replacement in patients with isolated MR (18, 36, 44).

In the present report, we tested whether desmin loss/degradation is an early finding in the pathophysiology of VO using TEM and immunohistochemistry and quantitated desmin cleavage with protein biochemistry. Here, we demonstrated increased desmin cleavage and disorganization of mitochondrial cytoskeletal organization starting as early as 24 h and continuing throughout the course of a pure VO by ACF to the 4- and 12-wk stages with an arrest of autophagy/mitophagy. Taken together, the pure stretch stimulus, neurohormonal activation, and cardiomyocyte ROS production support the notion that desmin cleavage is causal in the pathophysiological LV remodeling of a pure VO.

MATERIALS AND METHODS

Animal preparation.

Adult male Sprague-Dawley rats (200–250 g) at 12 wk of age were subjected to either sham or ACF surgery in our laboratory as previously described (9, 21, 45, 50, 54, 56). Briefly, rats were anesthetized under 2.5% isoflurane, and, under sterile conditions, an incision was made into the abdominal cavity whereby the descending aorta and vena cava were revealed. Blood flow was restricted through sufficient pressure to the main vessels in this region. A needle was first inserted into the aorta, and, while still inside the aorta, it was then pierced through into the vena cava. The needle was then retracted while the external hole from the initial aorta puncture was sealed with sterile gel adhesive. With the hole created between the major vessels, the pressure of aortic blood caused back flow into the vena cava leaving the fistula open, resulting in VO in the heart due to excess venous return. In control animals, a sham surgery procedure was conducted whereby all aspects of the surgery were performed except for the needle puncture to form the fistula. Time points of 24 h, 4 wk, and 12 wk were selected based on the development of heart failure from acute, compensated, and decompensated stages (9, 22). Rats that underwent sham or ACF surgery were studied at 4 or 12 wk after surgery using echocardiography before being euthanized, and whole LV tissue was used for protein analysis, TEM, and immunohistochemistry. Rats at 24 h or 4 or 12 wk after surgery were euthanized, and whole hearts used were to isolate LV cardiomyocytes. The animal use in these experiments was approved by the University of Alabama at Birmingham Animal Resource Program (protocol 130409070).

Echocardiography.

Echocardiography was performed before the animals were euthanized using the Vivo 2100 imaging system (Visualsonics, Toronto, ON, Canada) as previously described by our laboratory (22, 45, 54, 56). LV volume was calculated from traced M-mode LV dimensions using the following Teicholz formula: V = [7/(2.4 + LVID)] × [LVID]3, where V is volume and LVID is LV internal dimension.

Protein extraction from frozen tissue for desmin analysis.

For direct protein analysis, rats that underwent ACF surgery were dissected at 24 h, 4 wk, and 12 wk, and LV tissue was snap frozen in liquid nitrogen. Frozen tissue was pulverized using a liquid nitrogen-cooled mixer mill MM 400 (Retsch) and 0.5-mm cooled beads (2 × 1 min 30 s at 25 Hz). The resulting powder was resuspended in 25 mM HEPES buffer to separate myofilament- and cytosolic-enriched fractions as previously described (1).

Isolation of LV cardiomyocytes.

Cardiomyocytes were isolated from rats in both surgical groups as previously described by our laboratory (22, 45, 56). Briefly, hearts were perfused with perfusion buffer (120 mmol/l NaCl, 15 mmol/l KCl, 0.5 mmol/l KH2PO4, 5 mmol/l NaHCO3, 10 mmol/l HEPES, and 5 mmol/l glucose at pH 7.0) for 5 min and digested with perfusion buffer containing 1 mg/ml collagenase type II (Invitrogen, Carlsbad, CA) for 30 min at 37°C. The right ventricle, atria, and apex were removed before the perfused heart was minced. The digestion was filtered and washed, and cells were pelleted. Only samples with >80% viability (indicated by healthy rod-shaped cells) were used.

TEM of rat tissue.

Tissue was fixed in 2.5% glutaraldehyde-Sorensen’s phosphate buffer (no. 15980, Electron Microscopy Sciences, Hatfield, PA) overnight at 4°C as previously described by our laboratory (22, 50, 56). After postfixation with 1% osmium tetroxide in 0.1 M cacodylate buffer, tissue was dehydrated in a graded series of ethanol and embedded in Epon resin. Semithin (0.5 μm) and ultrathin (90 nm) sections were cut, mounted on copper grids, and poststained with uranyl acetate and lead citrate. Sections were viewed with a Philips 201 transmission electron microscope (FEI, Hillsboro, OR) for qualitative changes in ultrastructure by EMLabs (Birmingham, AL).

Immunohistochemistry.

Rat hearts were immersion fixed in 10% neutral-buffered formalin and embedded in paraffin. Sections (5 μm) were mounted on slides, deparaffinized in xylene, and rehydrated in a gradient series of ethanol. Sections used for fluorescent imaging were blocked with 5% goat serum and 1% bovine serum in PBS and incubated overnight at 4°C with either desmin antibody (1:100, ab6322, Abcam, Cambridge, UK) or voltage-dependent anion channel (VDAC; 1:100, ab15895, Abcam). Alexa fluor-conjugated secondary antibodies (1:500 each, Molecular Probes, Eugene, OR) were applied to visualize desmin (green) and VDAC (red) in the tissue. Nuclei (blue) were stained with DAPI (1.5 μg/ml, Vector Laboratories, Burlingame, CA). Image acquisition (×100 objective, ×4,000 video-screen magnification) was performed on a Leica DM6000 epifluorescence microscope with Simple-PCI software (Compix, Cranberry Township, PA). Images were adjusted appropriately to remove background fluorescence.

TUNEL assay.

A TUNEL assay was performed on paraffin-embedded sections using the DeadEnd fluorometric TUNEL kit (Promega, Madison, WI) as previously described by our laboratory (9).

Western blot analysis.

M-PER Mammalian Protein Extraction Reagent with Halt Phosphatase Inhibitor Cocktail and Halt Protease Inhibitor Cocktail (all from Life Technologies, Grand Island, NY) at 1:100 concentrations were used for our sample preparation. Cell lysates prepared from isolated cardiomyocytes (30–50 μg protein) were separated on a 4–12% bis-Tris gradient gel (Invitrogen) and transferred to a polyvinylidene difluoride (PVDF) membranes, which were incubated overnight at 4°C with the following antibodies: light chain (LC)3A/B (1:1,000, catalog no. 2741), p62/SQSTM1 (1:1,000, catalog no. 5114), beclin-1 (1:1,000, catalog no.3495), dynamin-related protein 1 (DRP1; 1:1,000, catalog no. 8570), phospho-DRP1 (Ser616, 1:1,000, catalog no. 3455), PTEN-induced putative kinase 1 (PINK1; 1:1,000, catalog no. 6946), parkin (1:1,000, catalog no. 4211), heat shock protein (HSP)60 (1:1,000, catalog no. 12165), Bad (1:1,000, catalog no. 9239), phospho-Bad (Ser155, 1:1,000, catalog no. 9297), Bcl-xL (1:1,000, catalog no. 2764), Akt (1:1,000, catalog no. 4691), and phospho-Akt (Ser473, 1:1,000, catalog no. 4060) (all from Cell Signaling Technology, Danvers, MA) as well as cyclophilin F (also known as CypD, 1:1,000, ab110324, Abcam) and GAPDH (1:1,000, GT239, GeneTex, Irvine, CA).

Membranes were then washed and incubated for 1 h with the appropriate Amersham enhanced chemiluminescence horseradish peroxidase-linked secondary antibody (GE Healthcare Bio-Sciences, Pittsburgh, PA). Membranes were saturated with chemiluminescent substrate (Pierce, Rockford, IL), and images were taken using a charge-coupled device camera and analyzed with ImageJ software (National Institutes of Health, Bethesda, MD). For desmin Western blot analysis on tissue, myofilament-enriched fractions were assayed for protein concentration using BCA (Pierce) diluted in lithium dodecyl sulfate buffer (Life Technologies) completed with 1% DTT and boiled at 95°C for 10 min. The resulting denatured samples were loaded onto 4–12% NuPAGE precast gels (Life Technologies), separated, and blotted on PVDF (Millipore) as previously described (1). Immunostaining using anti-desmin antibody (1:10,000 in Tris-buffered solution with 5% milk, DE-U-10; Sigma) was detected using a secondary red goat anti-mouse antibody (1:10,000, Li-Cor). After detection, membranes were stripped for 15 min (Restore, Pierce) and stained with Direct Blue 71 (Sigma) to confirm equal loading as previously described (1). For densitometry, the signal intensity of intact desmin was normalized to the average of sham-operated control animals at 24 h (equal to 100) to account for gel-to-gel variability. The signal intensity of the desmin fragment was expressed as a percentage of intact desmin within each lane, thus expressing the relative contribution of cleavage.

Mass spectrometry.

For mass spectrometry (MS) analysis, tissue protein extracts were separated on precast SDS-PAGE gels (bis-Tris 4–12%, Life Technologies) using MES buffer (Life Technologies). After separation, the gel slab was fixed overnight in 10% acetic acid-40% methanol (Fisher Scientific) followed by staining with blue silver staining (7). The electronic images of the desmin blot and the resulting Coomassie-stained gel were aligned using the molecular weight markers (SeeBlue; Life Technologies), and the corresponding bands were hand-picked using a clean scalpel and forceps. The excised bands were reduced to 1-mm3 cubes and subjected to in-gel digestion as previously described (1). Extracted peptides were analyzed using an Orbitrap Q Exactive plus MS (Thermo Scientific) with a 90-min gradient and 70K-35K resolution. The resulting spectra were searched against the Rattus norvegicus RefSeq2015 database with fixed carbamidomethyl, variable oxidation, deamidation, and phosphorylation with no enzyme (de novo sequencing) using Peaks (BSI).

Kinome analysis.

M-PER Mammalian Protein Extraction Reagent with Halt Phosphatase Inhibitor Cocktail and Halt Protease Inhibitor Cocktail (all from Life Technologies) at 1:100 concentrations were used for our sample preparation. Kinome analysis was performed at the University of Alabama Birmingham Kinome Core (Birmingham, AL). Before samples were loaded onto the four-well tyrosine (PTK) chips or four-well serine/threonine (STK) chips, lysates were quantified using a standard BCA assay. Lysates were loaded at 15 μg/well (PTK) or 2 μg/well (STK) onto the PamChips after preparation. The in vitro kinase assay was performed using PamGene Evolve software on the PamStation12. Images were then imported to BioNavigator for raw data analysis. As applicable upstream kinase prediction was performed with data from Kinexus Phosphonet, and pathway analysis was performed with GeneGo MetaCore.

Statistical analysis.

Values are presented as means ± SE. An unpaired t-test was used to compare age-matched rats in the sham-operated and ACF groups at each time point of 24 h, 4 wk, and 12 wk for Western blots, and a Mann-Whitney U-test using SPSS 24 software was used for echocardiography measures. P values of <0.05 were considered statistically significant.

RESULTS

Echocardiography.

A dimensional and volumetric echocardiographic analysis was performed to determine the extent of LV remodeling in response to the pure VO of ACF. As shown in Table 1, there were subtle increases in LV end-diastolic dimension and LV end-diastolic volume at 24 h that resulted in a significant decrease in the LV end-diastolic mass-to-volume ratio. As expected, there was an increase in LVEF and LV fractional shortening at this early stage of a pure stretch and ejection into the low-pressure fistula. At 4 and 12 wk of ACF, there was a significant increase in LV end-diastolic dimension (40% and 38%, respectively, P < 0.05) compared with age-matched sham-operated rats. However, there was a concomitant 140% and 130% increase in LV end-diastolic volume (P < 0.05). This reflects the change to a more spherical LV geometry manifested by a decrease in sphericity index at both 4 and 12 wk of ACF compared with age-matched sham-operated animals (1.75 ± 0.04 vs. 2.03 ± 0.07, P < 0.01, and 1.71 ± 0.02 vs. 1.87 ± 0.05, P < 0.05, respectively). Consistent with an eccentric and spherical remodeling pattern, there was a 48% and 56% increase in LV mass compared with the 2.5-fold increase in LV end-diastolic volume (both P < 0.05) and a decrease in the relative LV wall thickness at end diastole at 4 and 12 wk of ACF (P < 0.05). Indexes of systolic function, including LV fractional shortening, LVEF, and velocity of circumferential fractional shortening adjusted for heart rate (VCFr), did not differ from age-matched sham-operated animals at 4 wk of ACF; however, all were decreased significantly compared with age-matched sham-operated animals at 12 wk of ACF (P < 0.05). These echocardiographic indexes demonstrate that an adverse LV spherical eccentric remodeling at 4 and 12 wk of ACF ultimately results in a decrease in LV shortening indexes at 12 wk.

Table 1. Echocardiographic data

Measurement 24-h Sham 24-h ACF 4-wk Sham 4-wk ACF 12-wk Sham 12-wk ACF
n 8 8 11 12 9 8
Body weight, g 262 ± 8 275 ± 21 343 ± 6 359 ± 5* 472 ± 22 485 ± 26
HR, beats/min 408 ± 10 421 ± 13 348 ± 5 321 ± 5* 335 ± 18 322 ± 8
LVEDD, mm 6.71 ± 0.29 7.15 ± 0.16 7.32 ± 0.20 10.27 ± 0.19* 8.19 ± 0.15 11.27 ± 0.15*
LVESD, mm 3.94 ± 0.30 3.57 ± 0.19 4.71 ± 0.19 6.56 ± 0.15* 5.05 ± 0.11 7.73 ± 0.16*
LVPWTd, mm 1.88 ± 0.11 1.80 ± 0.06 1.88 ± 0.11 1.93 ± 0.13 1.93 ± 0.10 2.17 ± 0.14
LVPWTs, mm 2.79 ± 0.19 3.03 ± 0.10 2.98 ± 0.11 3.11 ± 0.15 2.90 ± 0.11 3.11 ± 0.13
RWT at diastole, 2 × pw/d 0.57 ± 0.05 0.51 ± 0.02 0.52 ± 0.05 0.39 ± 0.02* 0.47 ± 0.03 0.39 ± 0.03*
Sphericity index at diastole 2.02 ± 0.06 1.96 ± 0.06 2.03 ± 0.07 1.75 ± 0.04* 1.87 ± 0.05 1.71 ± 0.02*
Sphericity index at systole 2.98 ± 0.21 3.11 ± 0.17 2.69 ± 0.13 2.17 ± 0.10* 2.56 ± 0.05 2.18 ± 0.04*
LVESV, µl 111 ± 18 94 ± 10 149 ± 9 319 ± 26* 174 ± 18 532 ± 32*
LVEDV, µl 355 ± 35 409 ± 11 410 ± 19 975 ± 26* 560 ± 17 1282 ± 46*
LV mass, mg 759 ± 37 741 ± 21 665 ± 42 981 ± 61* 904 ± 71 1413 ± 149*
LV mass/LVEDV 2.35 ± 0.14 1.86 ± 0.10* 1.62 ± 0.07 1.01 ± 0.07* 1.61 ± 0.11 1.11 ± 0.12*
LVEF, % 71 ± 3 78 ± 2* 64 ± 2 67 ± 2 69 ± 3 59 ± 2*
LVFS, % 42 ± 2 48 ± 2* 40 ± 2 39 ± 1 40 ± 2 33 ± 1*
VCFr, % 8.7 ± 0.6 9.6 ± 0.4 7.4 ± 0.4 6.7 ± 0.3 7.4 ± 0.3 4.8 ± 0.1*

Values are means ± SE. The table shows dimensional and volumetric echocardiographic analysis of the extent of left ventricular (LV) remodeling in response to a pure volume overload of aortocaval fistula (ACF). HR, heart rate; LVEDD, LV end-diastolic dimension; LVESD, LV end-systolic dimension; LVPWTd, LV posterior wall thickness at diastole; LVPWTs, LV posterior wall thickness at systole; RWT, relative wall thickness; LVESV, LV end-systolic volume; LVEDV, LV end-diastolic volume; LVEF, LV ejection fraction; LV FS, LV fractional shortening; VCFr, velocity of circumferential fractional shortening adjusted for HR.

*P < 0.05, animals with ACF vs. age-matched sham-operated animals.

Desmin and mitochondrial loss of organization and function with ACF.

We recently reported a loss of organization in the cardiomyocyte cytoskeletal-mitochondrial architecture in rats with pure VO and in rats with ACF (56) and in humans with isolated MR (4). In the present study, we now demonstrate that desmin breakdown and mitochondrial disorganization occur as early as 24 h after ACF induction and persist at 4 and 12 wk of ACF (Fig. 1). In the normal LV, immmunofluorescent staining for desmin demonstrated a normal striated registry at the Z disks and more intense staining at the intercalated disks with a linear pattern of VDAC staining between sarcomeres. In hearts with ACF, there was near complete loss of desmin staining in some cardiomyocytes with intense VDAC staining that was consistent with the mitochondrial clustering in TEM images that characterize ACF sections (Fig. 2). The normal interfibrillar position of mitochondria closely straddling the A band of the sarcomere and its proximity to the myofibrillar structure were found in age-matched sham-operated and ACF rats, as shown in Fig. 2. In rats with ACF, at all time points, there was mitochondrial clustering with numerous small mitochondria that disrupted the normal highly organized linear registry of mitochondria with the sarcomere between the Z disks (Fig. 2). This disorganization was accompanied by the loss of the I bands and blurring of the Z disk. The mitochondrial disorganization at 24 h and 4 wk became more severe at 12 wk in animals with ACF with mitochondrial matrix swelling and cristae fragmentation, as demonstrated in the TEM images from two different rats with ACF shown in Fig. 3. It is of interest that these changes in mitochondrial disorganization and cristae loss along with myofibrillar breakdown were similar to those described in des/des−/− mice and in R120G-αβ-crystallin transgenic mice in very early stages before their early demise (37, 51, 52). These mitochondrial abnormalities were also consistent with our previous studies of ACF at 8 wk, in which there was a dissipation of mitochondrial membrane potential (ΔΨm), which was restored by the mitochondrial antioxidant MitoQ (56).

Fig. 1.

Fig. 1.Immunohistochemistry for desmin (green) and voltage-dependent anion channel (VDAC) (red) in the left ventricle (LV) of rats with aortocaval fistula (ACF) at 24 h, 4 wk, and 12 wk and age-matched sham-operated control animals. Images at each time point for sham-operated animals demonstrated normal desmin staining at the Z disk and intercalated disk associated with a linear distribution of VDAC staining for mitochondria. In the LV of animals with ACF, there was diffuse loss of desmin and VDAC staining in some myocytes and large areas of intense VDAC staining, consistent with the mitochondrial clustering in transmission electron microscopy (TEM) images.


Fig. 2.

Fig. 2.TEM images at ×4,500 of tissue with ACF at 24 h and 4 and 12 wk. TEM images demonstrated mitochondrial clustering with numerous small mitochondria in ACF cardiomyocytes. There was a disruption of the highly organized linear registry of mitochondria observed in sham-operated control animals, with more elongated mitochondria that straddle the A band of sarcomere in the ACF. M, mitochondria; N, nucleus; →, regions of mitochondrial disruption.


Fig. 3.

Fig. 3.TEM images of ACF at 12 wk. TEM images from LVs of one sham-operated rat at 12 wk (top) and two rats with ACF at 12 wk (middle and bottom) are shown. Left: magnification ×4,000; right: magnification ×12,000. White boxes delineate higher magnification areas. There was mitochondrial swelling with dissolution of cristae throughout the cardiomyocyte. M, mitochondria; N, nucleus.


Desmin cleavage and ACF and translocation of αB-crystallin.

Due to the profound changes observed in the desmin cytoskeleton, we investigated the idea that desmin cleavage could represent an early sign of adverse remodeling as manifested by the decrease in the LV end-diastolic volume mass-to-volume ratio. We measured the desmin cleavage product in ACF as early as 24 h after surgery using Western blot analysis (Fig. 4). Desmin and a 49-kDa band were separated using conventional SDS-PAGE. The 49-kDa fragment was approximately twofold increased at 24 h (P = 0.016) versus age-matched sham-operated animals, whereas intact desmin was only 10% increased (P = 0.04). At 4 wk of ACF, the 49-kDa fragment was increased approximately threefold (P = 0.021) with respect to age-matched sham-operated animals, whereas intact desmin was elevated by only 19% (P = 0.036). At 12 wk of ACF, there was no difference in intact desmin, whereas the 49-kDa fragment was increased twofold (P = 0.055). A second desmin fragment of ~25 kDa was also detected, but it did not display any significant difference at the three time points in animals with ACF versus sham-operated animals (Fig. 5, top).

Fig. 4.

Fig. 4.Western blot for desmin in LVs of rats with ACF at 24 h and 4 and 12 wk and in age-matched sham-operated rats. There was increased desmin degradation at 24 h and 4 wk in ACF samples by Western blot analysis, whereas there was no effect in the samples at 12 wk. Quantification (n = 7 per group) demonstrated increased desmin cleavage product (49-kDa protein at 24 h and 4 wk with ACF compared with age-matched sham-operated animals). Total intact desmin was also increased with ACF at both 24 h and 4 wk compared with age-matched sham-operated animals.


Fig. 5.

Fig. 5.Mass spectrometry (MS) sequence coverage of desmin and its fragments. Desmin sequence coverage was also confirmed by MS in the bands corresponding to intact desmin (53 kDa) and the fragments at ∼49 and ∼25 kDa (F1 and F2, respectively).


To rule out the possibility of nonspecific staining with the desmin antibody, we excised the corresponding bands from Coomassie-stained gels and subjected them to analysis by MS. The presence of desmin in all bands detected by the antibody was confirmed by de novo sequencing. Interestingly, the NH2-terminus of desmin, which is important for its assembly, was missing in both fragments (Fig. 5, bottom right).

Desmin’s chaperone protein, αB-crystallin, moved from its normal location at the Z disk (6) to the perinuclear area in rats with ACF at 4 and 12 wk. αB-crystallin followed the normal striated pattern along the Z disk in age-matched sham-operated control animals at 4 and 12 wk. In contrast, this regular distribution was replaced by αB-crystallin accumulation in the perinuclear area in ACF at both 4 and 12 wk (Fig. 6). These findings are consistent with what was also reported in patients with isolated MR and preserved LVEF (4).

Fig. 6.

Fig. 6.Immunohistochemistry for αB-crystallin (red) in LVs of rats with ACF at 4 and 12 wk and in age-matched sham-operated rats. In normal sham-operated animals, αB-crystallin (red) was colocalized to the Z disk. In LVs with ACF, there was extensive desmin loss along with areas of aggregation of αβ-crystallin most prominent in the perinuclear region.


Oxidative stress in cardiomyocytes.

One of the major causes of desmin disruption and mitochondrial disarray is excessive oxidative stress. We have previously demonstrated increased oxidative stress in isolated cardiomyocytes with ACF at 24 h (50) and 8 wk (56). Here, we studied the distribution of hydroxynonenal (HNE; Fig. 7, red) (4), which is a strong indicator of lipid peroxidation. As we have previously reported in people with isolated MR (4), there was a marked increase in HNE intensity in cardiomyocytes with ACF at both the 4- and 12-wk time points compared with age-matched sham-operated control animals. HNE staining was especially prominent in the perinuclear region, which coincides with damaged mitochondria in the TEM images shown in Fig. 2 and VDAC staining shown in Fig. 1 in hearts with ACF. The colocalization of HNE and VDAC is consistent with lipid peroxidation of the membrane-rich mitochondria in the cardiomyocyte.

Fig. 7.

Fig. 7.Immunohistochemistry for VDAC (green) and 4-hydroxy-2-nonenal (HNE; red) in LVs with ACF and those from sham-operated animals. The sham-operated LV had a striated, linear pattern of VDAC (green) with very little HNE staining (red). Blue shows nuclei stained with DAPI. In hearts with ACF, there was extensive VDAC loss and perinuclear increased staining along with increased HNE staining that corresponded with the mitochondrial clustering and sarcomeric breakdown in the TEM images.


Autophagy and apoptosis.

The marked loss of organization of desmin cytoskeleton and mitochondria as visualized with TEM has been associated with an arrest of autophagy in the des/des−/− mouse. In the cardiomyocytes with ACF at 4 and 12 wk that we studied, there was a significant increase in several autophagy protein markers, including microtubule-associated protein 1A/1B-LC3 (LC3-II/LC3-I ratio), p62/SQSTM1, and beclin-1 (Fig. 8A). In addition, LC3-II/GAPDH was increased 2.24- and 1.98-fold compared with age-matched sham-operated rats and those with ACF at both 4 and 12 wk, respectively.

Fig. 8.

Fig. 8.Upregulation of autophagy and antiapoptotic protein markers in LV cardiomyocytes of animals with ACF at 4 and 12 wk. Cardiomyocytes were isolated from animals with ACF at 4 wk and 12 wk and from sham-operated rats. Equal amounts of cell lysate were analyzed by SDS-PAGE and transferred to a polyvinylidene difluoride membrane followed by immunoblot analysis. A: specific antibodies for light chain (LC)3, p62, beclin, and GAPDH. Bar graphs at right show fold increases in cardiomyocytes from animals with ACF at 4 and 12 wk compared with sham-operated animals for LC3II/GAPD, p62, and beclin-1 (n = 4 for each group, *P < 0.05). B: representative images of Western blots for mitochondrial damage and mitophagy markers in age-matched sham-operated rats and rats with ACF at 4 and 12 wk. Bar graphs at right show significant fold increases in phosphorylated (p-)dynamin-related protein 1 (DRP1), DRP1, PTEN-induced putative kinase 1 (PINK1), parkin, heat shock protein (Hsp)60, and cyclophilin D (CypD) (after being normalized to GAPDH) in cardiomyocytes of animals with ACF at 4 and 12 wk compared with sham-operated animals (n = 4 for each group, *P < 0.05).


Markers of mitophagy were also increased with ACF at 4 and 12 wk, including total DRP1 and Ser616-phosphorylated phospho-DRP1, PINK1, parkin, HSP60, and CypD (Fig. 8B). DRP1 affects mitochondrial morphology and is important in mitochondrial fission in mammalian cells. Specifically, DRP1 phosphorylation at Ser616 promotes mitochondrial fission (17), which has been reported to precede mitophagy and make mitochondria more suitable for engulfment by the autophagic machinery (39). However, this increase in mitophagy protein markers was countered by a significant sixfold increase in p62/SQSTM1 (Fig. 8A), which accumulates when autophagy is inhibited and decreases when autophagy is induced (30). Increased mitophagy markers may be affected by differences in total mitochondrial abundance, but this was ruled out by measured citrate synthase activity, which was not significantly different between sham-operated control animals and those with ACF at 12 wk (data not shown). Nevertheless, these are static markers of mitophagy in the face of extensive desmin/mitochondrial damage. Thus, it is possible that autophagy/mitophagy is activated and thereby increases the autophagic/mitophagic flux, but this increase may still not be adequate to remove the damaged mitochondria in a timely fashion at these stages of ACF VO. In addition, apoptosis was not induced by ACF at either 4 or 12 wk, as indicated both by increased protein levels of the antiapoptotic markers including total Akt, phospho-Akt (Ser473), total Bad, phospho-Bad (Ser155), and Bcl-xL (Fig. 9) and by TUNEL staining (Fig. 10).

Fig. 9.

Fig. 9.Upregulation of antiapoptotic protein markers in cardiomyocytes of rats with ACF. A: cardiomyocyte lysates were prepared as described in Fig. 7. Representative Western blots are shown for rats with ACF at 4 and 12 wk and for age-matched sham-operated control rats. B: bar graph showing the significant fold increases in Bad, p-Bad/Bad, Bcl-xL, Akt, and p-Akt/Akt at 4 wk of ACF compared with levels in sham-operated animals. C: bar graph showing significant fold increases in Bad, p-Bad/Bad, Bcl-xL, Akt, and p-Akt/Akt (normalized to GAPDH) in cardiomyocytes of animals with ACF at 12 wk compared with sham-operated animals. n = 4 for each group. *P < 0.05.


Fig. 10.

Fig. 10.TUNEL staining demonstrates the absence of cardiomyocyte apoptosis in ACF. A TUNEL assay was performed on paraffin-embedded sections using the DeadEnd fluorometric TUNEL kit. Few to no LV cardiomyocytes showed positive TUNEL staining in animals with ACF at 4 and 12 wk compared with age-matched sham-operated animals. DNase I treatment of paraffin-embedded tissue sections, resulting in fragmentation of the chromosomal DNA, served as positive controls (green). Representative images of n = 3 each of sham, 4-wk ACF, and 12-wk ACF are shown. Red, laminin; blue, DAPI.


Kinome analysis.

To further interrogate and verify the mechanistic underpinnings of this antiapoptotic phenotype of the ACF cardiomyocyte by Western blot analysis, we performed a kinome analysis of LV cardiomyoctyes isolated from the LVs of rats with ACF at 4 and 12 wk and age-matched sham-operated rats. Kinomic profiling is a functional proteomic approach that can help elucidate the central intracellular signaling pathways driving particular biological processes or phenotypes (Tables 2 and 3) (16, 29).

Table 2. ACF kinome activation at 4 and 12 wk

Kinase UniProt Score
Serine/threonine kinase at 4 wk
    MSK1 (RPS6KA5) O75582 6360
    MAPKAPK3 Q16644 5292
    PKACb (PRKACB) P22694 5373
    Pim3 (AL549548) P58750 4975
    Pim2 Q9P1W9 4231
    PKG1 (PRKG1) Q13976 4025
    MAPKAPK2 P49137 4363
    Pim1 P11309 4155
    CaMK4 Q16566 2094
Serine/threonine kinase at 12 wk
    PKG1 (PRKG1) Q13976 1579.5
    PKACa (PRKACA) P17612 1550.5
    PKACb (PRKACB) P22694 1550.5
    PIM1 P11309 1332.5
    PKG2 (PRKG2) Q13237 1094
    AKT2 P31751 1033
    AKT1 P31749 1027
    AXL* P30530 979
    MSK1 (RPS6KA5) O75582 811.5
    Pim3 (AL549548) P58750 764.5
    PRKX P51817 682.5

The table shows upstream kinases and scores with verified cardiomyocyte expression for ACF at 4 and 12 wk. Comparisons of kinomic profiles between samples were performed using BioNavigator software version 5 (PamGene) to identify significantly different phosphopeptides. Using two proprietary algorithms, the upstream kinases were identified by scoring potential kinases based on their prevalence in the top 10 kinase scoring lists for each phosphopeptide as mapped in the Kinexus upstream kinase database (www.phosphonet.ca). The score is based on the number of phosphopeptides, and these scores are used to rank a kinase. Kinexus rankings are primarily known as algorithm-based interactions, not necessarily literature derived interactions, and thus a limitation of this analysis (16, 29). Of note, no kinases were less active in rats with ACF at 12 wk compared with rats at 12 wk in the sham-operated control group.

*AXL is a tyrosine kinase.

Table 3. Upstream kinases, identifiers, and biological function identified in kinome analysis

Kinase UniProtKB Full Name Biological Function
AKT1 P31749 Protein kinase B AGC protein kinase involved as one of three closely related serine/threonine protein kinases (AKT1, AKT2, and AKT3) called AKT kinase, which regulate many processes including metabolism, proliferation, cell survival, growth, and angiogenesis
AKT2 P31751 Protein kinase B-β
AXL P30530 Tyrosine protein kinase receptor UFO Axl family receptor tyrosine kinase involved in cell survival by preventing apoptosis, cell proliferation, migration, and differentiation
CaMK4 Q16566 Ca2+/calmodulin-dependent protein kinase type IV Ca2+/calmodulin-dependent protein kinase involved in transcriptional regulation, microtubule dynamics, and mitochondrial organization and biogenesis
MAPKAPK2 P49137 Mitogen-activated protein kinase activated kinase 2 MAPKAPK protein kinase involved in signaling in both mitogen and stress responses
MAPKAPK3 Q16644 Mitogen-activated protein kinase activated kinase 3
MSK1 O75582 90-kDa ribosomal protein S6 kinase 5 AGC protein kinase involved in the stress-activated MAPK cascade in response to growth factors and cellular stress
Pim1 P11309 Serine/threonine protein kinase Pim-1 Ca2+/calmodulin-dependent protein kinase involved in negative regulation of apoptosis and regulates cell cycle
Pim2 Q9P1W9 Serine/threonine protein kinase Pim-2
Pim3 P58750 Serine/threonine protein kinase Pim-3
PKACa P17612 cAMP-dependent protein kinase catalytic α-subunit AGC protein kinase involved in mediating cAMP-dependent signaling triggered by receptor binding to G protein-coupled receptors and regulates diverse cellular processes such as cell proliferation and differentiation and regulation of microtubule dynamics
PKACb P22694 cAMP-dependent protein kinase catalytic β-subunit
PKG1 Q13976 cGMP-dependent protein kinase 1 AGC protein kinase involved in cell signaling, gene expression, and nitric oxide formation
PKG2 Q13237 cGMP-dependent protein kinase 2
PRKX P51817 Protein kinase, X linked AGC protein kinase involved in multiple functions in cellular differentiation and epithelial morphogenesis including angiogenesis

The tyrosine kinome at 4 wk of ACF did not show any significant differences from that of control animals at 4 wk. The serine/threonine kinome identified activation of MSK1 (90-kDa ribosomal protein S6 kinase 5) involved in the stress-activated MAPK cascade in response to cellular stress and inflammation; MAPK-activated protein kinases 2 and 3 (MAPKAPK2 and MAPKAPK3, respectively) involved in cell growth, tissue-specific gene expression, and cell differentiation in response to cellular stress; 70-kDa ribosomal protein S6 kinase 2 (p70S6Kb) involved in translational initiation and cell growth; and Ca2+/calmodulin-dependent protein kinase IV (CaMK4) involved in transcriptional regulation and mitochondrial organization and biogenesis. There was an induction of PIM1, PIM2, and PIM3 protooncogene serine/threonine-protein kinases involved in cell survival and proliferation and negative regulation of apoptosis by inhibition of proapoptotic proteins.

The tyrosine kinome at 12 wk of ACF had activation of AXL, an antiapoptotic receptor tyrosine kinase involved in cell growth (5), and the serine/threonine kinome had sustained activation of PIM1 and PIM3 as well as PKB (Akt1 and Akt2), which play a critical role in regulating cell survival and metabolism via many different signaling pathways (20). The serine/threonine kinome also showed activation of PKG (PKG1 and PKG2), which is involved in actin cytoskeleton organization and biogenesis, and PKA (PKACa and PKACb), which is involved in actin dynamics, microtubule dynamics, and inhibition of apoptosis. With regard to potential posttranslation modifications of desmin (28, 49, 55), the serine/threonine kinome demonstrated activation of PKG1, PKG2, PKACa, and PKACb (Tables 2 and 3).

DISCUSSION

In the present study, we provide further insights into our previous findings of increased desmin breakdown and cleavage in rats with chronic ACF (56) and in humans with isolated chronic MR and LVEF of >60% (4). The breakdown of desmin after just 24 h of ACF is associated with disruption of the highly ordered mitochondrial sarcomeric lattice architecture. As in the human heart with isolated MR, mitochondrial damage was associated with increased HNE and perinuclear accumulation of αB-crystallin at compensated (4 wk) and late stages (12 wk) of ACF. Desmin cleavage increased two- to threefold at 24 h of ACF and at 4 wk of ACF, whereas intact desmin was only slightly increased throughout ACF. The early onset of desmin cleavage and mitochondrial disorganization suggests that cytoskeletal breakdown may be causative in LV dilatation and spherical remodeling in the progression to heart failure in a pure VO.

Desmin is the major protein component of cardiac intermediate filaments, a highly abundant and yet understudied portion of the cytoskeleton (31, 49), in the pathophysiology of nongenetic causes of heart failure. The canonical view is that intermediate filaments maintain the relative positioning of different organelles and cell structures in the heart. However, this “static” view has been challenged by a number of reports that have described dynamic functions for desmin in regulating mitochondrial function in transgenic models (15) and force development in cardiomyoctyes (33, 41). Mice carrying R120G, a mutated form of αB-crystallin (46), the major chaperone of desmin, quickly develop desmin-related dilated cardiomyopathy and mitochondrial abnormalities (32, 52). These abnormalities are accompanied by the deposition of desmin aggregates whereby the cellular autophagy process is insufficient to resolve their pathogenic accumulation, leading to the onset of a dilated cardiomyopathy. The phenotype of this well-established transgenic model of cardiac proteotoxicity can be rescued by increased autophagy (42) or increased expression of chaperone proteins (15, 47). Desmin misarrangement and mitochondrial disorganization at 24 h in the hearts with VO that we studied are similar to those described in both the αB-crystallin and des/des−/− transgenic mice in early stages before their demise from dilated cardiomyopathy (37, 51).

In addition to an early disorganized clustering of small interfibrillar and subsarcolemmal mitochondria, there is a loss of cristae and breakdown of perinuclear mitochondria in cardiomyocytes with ACF at 12 wk and a lack of apoptosis by protein analysis that was supported by a screening kinome analysis. Protein analysis also suggests an upregulation of mitophagy to clear morphologically damaged mitochondria. However, there is a relative absence of extensive mitophagy by TEM analysis and a failure of autophagic clearance of desmin by its major chaperone as manifested by a perinuclear accumulation of αB-crystallin. The failure of a completion of mitophagy/autophagy is further supported by the marked increase of p62 in cardiomyocytes with ACF at 4 and 12 wk; however, alternatively, it should be considered, as was observed in the R120G-CryAB cardiomyopathy model, that increased p62 can coincide with increased autophagy (59). Thus, the present study cannot characterize whether the progressive pathology of the cardiomyocyte in VO may be due to diminished autophagic response or an insufficient stress-induced upregulation in mitophagy to handle the aggregate burden of extensive desmin degradation and mitochondrial damage. Desmin loss of function due to cleavage and its tight connection to mitochondria should also be considered. In fact, accumulation of damaged mitochondria can be a source of persistent ROS production, creating a vicious cycle that can predispose to desmin cleavage and other posttranslational modifications (2, 55).

Our previous studies of cyclical stretch in adult rat cardiomyocytes (22, 56) support the contention that mitochondrial ROS production is an important driver of the cytoskeletal disruption in VO. In these studies, stretch caused mitochondrial ROS production that resulted in breakdown of the Z disk and myofibrils, which was prevented by pretreatment with the mitochondrial antioxidant MitoQ or xanthine oxidase inhibitor allopurinol. The apparent patchy loss of desmin staining in ACF cardiomyocytes by immunohistochemistry without a decrease in desmin protein by Western blot analysis may result from numerous potential desmin posttranslational modifications that can obscure the epitope from antibody recognition (14, 55) but nonetheless result in blurring of the Z disk by TEM at all stages of ACF.

Although it is beyond the scope of this study, many posttranslational modifications besides cleavage have been implicated in desmin-mitochondrial structural pathology, including phosphorylation, oxidation, and glycation (55). Our cardiomyocyte kinome analysis at the 4- and 12-wk stages (Table 2) identifies potential kinases involved in desmin phosphorylation in disease states (55); in particular, PKAs that result from excessive adrenergic drive, which is well documented in VO (24, 26, 35, 38, 53, 54, 58). Desmin surrounds and interlinks the Z disks and forms a lattice that connect the myofibrils, sarcolemma, mitochondria, sarcoplasmic reticulum, and nuclei. The Z disks have been described not only as a nodal point for mechanosensation (19) but also as the location of kinases, phosphatases, and Ca2+ signaling molecules. Specifically, PKC and PKA have been shown to translocate to anchoring proteins in the Z disk after activation (43). Thus, stretch, activation, and translocation of G protein-coupled receptor-activated kinases provide a cellular mechanism for the phosphorylation of desmin at the Z disk and the subsequent desmin changes that can cause deranged mitochondrial sarcomeric architecture as in the hearts with VO that we studied.

The present study demonstrates that in a pure VO there is a continuous desmin misarrangement and disruption of the normal mitochondrial sarcomeric architecture that begins as early as 24 h, suggesting a causal link between desmin cleavage/alterations and mitochondrial disorganization and damage. Desmin is a major substrate for the intracellular Ca2+-dependent protease calpain (23, 41), which is activated during the high levels of free intracellular Ca2+ achieved during ischemia-reperfusion (41) but also with increased TNF-α (40) and excessive β-adrenergic signaling and oxidative stress (33, 41). Taken together, the early and incessant adrenergic drive and cardiomyocyte oxidative stress of pure VO may be the link to protease activation and desmin cleavage and other posttranslational modifications that start an adverse remodeling of misarrangement of desmin at the Z disk. Due to the established link between mitochondrial function and desmin, this could result in a self-sustained maladaptive feedback loop. Future studies will attempt to unravel the cause and effect of posttranslational modifications of desmin that predispose it to cleavage and/or potential conformation changes that initiate a cascade of events driving mitochondria damage and ROS production, myofibrillar disruption, decreased force development, and insufficient autophagy/mitophagy responses.

GRANTS

Support for this study was provided by Department of Veterans Affairs Merit Fund Grant BX003664-01 and National Institutes of Health (NIH) Grant P01 HL-107153 (to L. J. Dell’Italia), NIH Training Grant 5-T32-HL-072757 (to J. L. Guichard), NIH Interdisciplinary Training in Pathobiology and Rehabilitation Medicine Grant 1-T32-HD-071866 (to M. P. Rogowski), American Heart Association Grants 2SDG9210000 and 16IRG27240002, the Magic that Matters Foundation (Johns Hopkins University SOM), and by RFO University of Bologna support (to G. Agnetti).

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

J.G. and L.J.D. conceived and designed research; J.G., M.P.R., G.A., L.F., P.P., and C.-C.W. performed experiments; J.G., M.P.R., G.A., L.F., P.P., and C.-C.W. analyzed data; J.G., M.P.R., G.A., L.F., C.-C.W., J.F.C., and L.J.D. interpreted results of experiments; J.G., M.P.R., G.A., L.F., and P.P. prepared figures; J.G., M.P.R., and L.J.D. drafted manuscript; J.G., M.P.R., G.A., J.F.C., and L.J.D. edited and revised manuscript; M.P.R., J.F.C., and L.J.D. approved final version of manuscript.

REFERENCES

  • 1. Agnetti G, Halperin VL, Kirk JA, Chakir K, Guo Y, Lund L, Nicolini F, Gherli T, Guarnieri C, Caldarera CM, Tomaselli GF, Kass DA, Van Eyk JE. Desmin modifications associate with amyloid-like oligomers deposition in heart failure. Cardiovasc Res 102: 24–34, 2014. doi:10.1093/cvr/cvu003.
    Crossref | PubMed | Web of Science | Google Scholar
  • 2. Agnetti G, Husberg C, Van Eyk JE. Divide and conquer: the application of organelle proteomics to heart failure. Circ Res 108: 512–526, 2011. doi:10.1161/CIRCRESAHA.110.226910.
    Crossref | PubMed | Web of Science | Google Scholar
  • 3. Ahmed MI, Gladden JD, Litovsky SH, Lloyd SG, Gupta H, Inusah S, Denney T Jr, Powell P, McGiffin DC, Dell’Italia LJ. Increased oxidative stress and cardiomyocyte myofibrillar degeneration in patients with chronic isolated mitral regurgitation and ejection fraction >60%. J Am Coll Cardiol 55: 671–679, 2010. doi:10.1016/j.jacc.2009.08.074.
    Crossref | PubMed | Web of Science | Google Scholar
  • 4. Ahmed MI, Guichard JL, Rajasekaran NS, Ahmad S, Mariappan N, Litovsky S, Gupta H, Lloyd SG, Denney TS, Powell PC, Aban I, Collawn JF, Davies JE, McGiffin DC, Dell’Italia LJ. Disruption of desmin-mitochondrial architecture in patients with regurgitant mitral valves and preserved ventricular function. J Thorac Cardiovasc Surg 152: 1059–1070.e2, 2016. doi:10.1016/j.jtcvs.2016.06.017.
    Crossref | PubMed | Web of Science | Google Scholar
  • 5. Bellosta P, Zhang Q, Goff SP, Basilico C. Signaling through the ARK tyrosine kinase receptor protects from apoptosis in the absence of growth stimulation. Oncogene 15: 2387–2397, 1997. doi:10.1038/sj.onc.1201419.
    Crossref | PubMed | Web of Science | Google Scholar
  • 6. Bennardini F, Wrzosek A, Chiesi M. Alpha B-crystallin in cardiac tissue. Association with actin and desmin filaments. Circ Res 71: 288–294, 1992. doi:10.1161/01.RES.71.2.288.
    Crossref | PubMed | Web of Science | Google Scholar
  • 7. Candiano G, Bruschi M, Musante L, Santucci L, Ghiggeri GM, Carnemolla B, Orecchia P, Zardi L, Righetti PG. Blue silver: a very sensitive colloidal Coomassie G-250 staining for proteome analysis. Electrophoresis 25: 1327–1333, 2004. doi:10.1002/elps.200305844.
    Crossref | PubMed | Web of Science | Google Scholar
  • 8. Capetanaki Y, Papathanasiou S, Diokmetzidou A, Vatsellas G, Tsikitis M. Desmin related disease: a matter of cell survival failure. Curr Opin Cell Biol 32: 113–120, 2015. doi:10.1016/j.ceb.2015.01.004.
    Crossref | PubMed | Web of Science | Google Scholar
  • 9. Chen YW, Pat B, Gladden JD, Zheng J, Powell P, Wei CC, Cui X, Husain A, Dell’Italia LJ. Dynamic molecular and histopathological changes in the extracellular matrix and inflammation in the transition to heart failure in isolated volume overload. Am J Physiol Heart Circ Physiol 300: H2251–H2260, 2011. doi:10.1152/ajpheart.01104.2010.
    Link | Web of Science | Google Scholar
  • 10. Dalakas MC, Park KY, Semino-Mora C, Lee HS, Sivakumar K, Goldfarb LG. Desmin myopathy, a skeletal myopathy with cardiomyopathy caused by mutations in the desmin gene. N Engl J Med 342: 770–780, 2000. doi:10.1056/NEJM200003163421104.
    Crossref | PubMed | Web of Science | Google Scholar
  • 11. Dell’italia LJ, Balcells E, Meng QC, Su X, Schultz D, Bishop SP, Machida N, Straeter-Knowlen IM, Hankes GH, Dillon R, Cartee RE, Oparil S. Volume-overload cardiac hypertrophy is unaffected by ACE inhibitor treatment in dogs. Am J Physiol 273: H961–H970, 1997.
    Link | Web of Science | Google Scholar
  • 12. Di Somma S, Di Benedetto MP, Salvatore G, Agozzino L, Ferranti F, Esposito S, La Dogana P, Scarano MI, Caputo G, Cotrufo M, Santo LD, de Divitiis O. Desmin-free cardiomyocytes and myocardial dysfunction in end stage heart failure. Eur J Heart Fail 6: 389–398, 2004. doi:10.1016/j.ejheart.2003.07.010.
    Crossref | PubMed | Web of Science | Google Scholar
  • 13. Di Somma S, Marotta M, Salvatore G, Cudemo G, Cuda G, De Vivo F, Di Benedetto MP, Ciaramella F, Caputo G, de Divitiis O. Changes in myocardial cytoskeletal intermediate filaments and myocyte contractile dysfunction in dilated cardiomyopathy: an in vivo study in humans. Heart 84: 659–667, 2000. doi:10.1136/heart.84.6.659.
    Crossref | PubMed | Web of Science | Google Scholar
  • 14. Diguet N, Mallat Y, Ladouce R, Clodic G, Prola A, Tritsch E, Blanc J, Larcher JC, Delcayre C, Samuel JL, Friguet B, Bolbach G, Li Z, Mericskay M. Muscle creatine kinase deficiency triggers both actin depolymerization and desmin disorganization by advanced glycation end products in dilated cardiomyopathy. J Biol Chem 286: 35007–35019, 2011. doi:10.1074/jbc.M111.252395.
    Crossref | PubMed | Web of Science | Google Scholar
  • 15. Diokmetzidou A, Soumaka E, Kloukina I, Tsikitis M, Makridakis M, Varela A, Davos CH, Georgopoulos S, Anesti V, Vlahou A, Capetanaki Y. Desmin and αB-crystallin interplay in the maintenance of mitochondrial homeostasis and cardiomyocyte survival. J Cell Sci 129: 3705–3720, 2016. doi:10.1242/jcs.192203.
    Crossref | PubMed | Web of Science | Google Scholar
  • 16. Duverger A, Wolschendorf F, Anderson JC, Wagner F, Bosque A, Shishido T, Jones J, Planelles V, Willey C, Cron RQ, Kutsch O. Kinase control of latent HIV-1 infection: PIM-1 kinase as a major contributor to HIV-1 reactivation. J Virol 88: 364–376, 2014. doi:10.1128/JVI.02682-13.
    Crossref | PubMed | Web of Science | Google Scholar
  • 17. Elgass K, Pakay J, Ryan MT, Palmer CS. Recent advances into the understanding of mitochondrial fission. Biochim Biophys Acta 1833: 150–161, 2013. doi:10.1016/j.bbamcr.2012.05.002.
    Crossref | PubMed | Web of Science | Google Scholar
  • 18. Enriquez-Sarano M, Suri RM, Clavel MA, Mantovani F, Michelena HI, Pislaru S, Mahoney DW, Schaff HV. Is there an outcome penalty linked to guideline-based indications for valvular surgery? Early and long-term analysis of patients with organic mitral regurgitation. J Thorac Cardiovasc Surg 150: 50–58, 2015. doi:10.1016/j.jtcvs.2015.04.009.
    Crossref | PubMed | Web of Science | Google Scholar
  • 19. Frank D, Frey N. Cardiac Z-disc signaling network. J Biol Chem 286: 9897–9904, 2011. doi:10.1074/jbc.R110.174268.
    Crossref | PubMed | Web of Science | Google Scholar
  • 20. Franke TF, Kaplan DR, Cantley LC. PI3K: downstream AKTion blocks apoptosis. Cell 88: 435–437, 1997. doi:10.1016/S0092-8674(00)81883-8.
    Crossref | PubMed | Web of Science | Google Scholar
  • 21. Gladden JD, Zelickson BR, Guichard JL, Ahmed MI, Yancey DM, Ballinger S, Shanmugam M, Babu GJ, Johnson MS, Darley-Usmar V, Dell’Italia LJ. Xanthine oxidase inhibition preserves left ventricular systolic but not diastolic function in cardiac volume overload. Am J Physiol Heart Circ Physiol 305: H1440–H1450, 2013. doi:10.1152/ajpheart.00007.2013.
    Link | Web of Science | Google Scholar
  • 22. Gladden JD, Zelickson BR, Wei CC, Ulasova E, Zheng J, Ahmed MI, Chen Y, Bamman M, Ballinger S, Darley-Usmar V, Dell’Italia LJ. Novel insights into interactions between mitochondria and xanthine oxidase in acute cardiac volume overload. Free Radic Biol Med 51: 1975–1984, 2011. doi:10.1016/j.freeradbiomed.2011.08.022.
    Crossref | PubMed | Web of Science | Google Scholar
  • 23. Goll DE, Thompson VF, Li H, Wei W, Cong J. The calpain system. Physiol Rev 83: 731–801, 2003. doi:10.1152/physrev.00029.2002.
    Link | Web of Science | Google Scholar
  • 24. Grossman PM, Linares OA, Supiano MA, Oral H, Mehta RH, Starling MR. Cardiac-specific norepinephrine mass transport and its relationship to left ventricular size and systolic performance. Am J Physiol Heart Circ Physiol 287: H878–H888, 2004. doi:10.1152/ajpheart.00007.2003.
    Link | Web of Science | Google Scholar
  • 25. Grossman W, Jones D, McLaurin LP. Wall stress and patterns of hypertrophy in the human left ventricle. J Clin Invest 56: 56–64, 1975. doi:10.1172/JCI108079.
    Crossref | PubMed | Web of Science | Google Scholar
  • 26. Hankes GH, Ardell JL, Tallaj J, Wei CC, Aban I, Holland M, Rynders P, Dillon R, Cardinal R, Hoover DB, Armour JA, Husain A, Dell’Italia LJ. β1-Adrenoceptor blockade mitigates excessive norepinephrine release into cardiac interstitium in mitral regurgitation in dog. Am J Physiol Heart Circ Physiol 291: H147–H151, 2006. doi:10.1152/ajpheart.00951.2005.
    Link | Web of Science | Google Scholar
  • 27. Hnia K, Ramspacher C, Vermot J, Laporte J. Desmin in muscle and associated diseases: beyond the structural function. Cell Tissue Res 360: 591–608, 2015. doi:10.1007/s00441-014-2016-4.
    Crossref | PubMed | Web of Science | Google Scholar
  • 28. Huang X, Li J, Foster D, Lemanski SL, Dube DK, Zhang C, Lemanski LF. Protein kinase C-mediated desmin phosphorylation is related to myofibril disarray in cardiomyopathic hamster heart. Exp Biol Med (Maywood) 227: 1039–1046, 2002.
    Crossref | PubMed | Web of Science | Google Scholar
  • 29. Jarboe JS, Jaboin JJ, Anderson JC, Nowsheen S, Stanley JA, Naji F, Ruijtenbeek R, Tu T, Hallahan DE, Yang ES, Bonner JA, Willey CD. Kinomic profiling approach identifies Trk as a novel radiation modulator. Radiother Oncol 103: 380–387, 2012. doi:10.1016/j.radonc.2012.03.014.
    Crossref | PubMed | Web of Science | Google Scholar
  • 30. Komatsu M, Ichimura Y. Physiological significance of selective degradation of p62 by autophagy. FEBS Lett 584: 1374–1378, 2010. doi:10.1016/j.febslet.2010.02.017.
    Crossref | PubMed | Web of Science | Google Scholar
  • 31. Lazarides E. Intermediate filaments as mechanical integrators of cellular space. Nature 283: 249–256, 1980. doi:10.1038/283249a0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 32. Maloyan A, Gulick J, Glabe CG, Kayed R, Robbins J. Exercise reverses preamyloid oligomer and prolongs survival in αB-crystallin-based desmin-related cardiomyopathy. Proc Natl Acad Sci USA 104: 5995–6000, 2007. doi:10.1073/pnas.0609202104.
    Crossref | PubMed | Web of Science | Google Scholar
  • 33. Matsumura Y, Saeki E, Inoue M, Hori M, Kamada T, Kusuoka H. Inhomogeneous disappearance of myofilament-related cytoskeletal proteins in stunned myocardium of guinea pig. Circ Res 79: 447–454, 1996. doi:10.1161/01.RES.79.3.447.
    Crossref | PubMed | Web of Science | Google Scholar
  • 34. McLendon PM, Robbins J. Desmin-related cardiomyopathy: an unfolding story. Am J Physiol Heart Circ Physiol 301: H1220–H1228, 2011. doi:10.1152/ajpheart.00601.2011.
    Link | Web of Science | Google Scholar
  • 35. Mehta RH, Supiano MA, Oral H, Grossman PM, Montgomery DS, Smith MJ, Starling MR. Compared with control subjects, the systemic sympathetic nervous system is activated in patients with mitral regurgitation. Am Heart J 145: 1078–1085, 2003. doi:10.1016/S0002-8703(03)00111-X.
    Crossref | PubMed | Web of Science | Google Scholar
  • 36. Miller JD, Suri RM. Left ventricular dysfunction after degenerative mitral valve repair: A question of better molecular targets or better surgical timing? J Thorac Cardiovasc Surg 152: 1071–1074, 2016. doi:10.1016/j.jtcvs.2016.07.018.
    Crossref | PubMed | Web of Science | Google Scholar
  • 37. Milner DJ, Weitzer G, Tran D, Bradley A, Capetanaki Y. Disruption of muscle architecture and myocardial degeneration in mice lacking desmin. J Cell Biol 134: 1255–1270, 1996. doi:10.1083/jcb.134.5.1255.
    Crossref | PubMed | Web of Science | Google Scholar
  • 38. Nagatsu M, Zile MR, Tsutsui H, Schmid PG, DeFreyte G, Cooper G IV, Carabello BA. Native beta-adrenergic support for left ventricular dysfunction in experimental mitral regurgitation normalizes indexes of pump and contractile function. Circulation 89: 818–826, 1994. doi:10.1161/01.CIR.89.2.818.
    Crossref | PubMed | Web of Science | Google Scholar
  • 39. Novak I. Mitophagy: a complex mechanism of mitochondrial removal. Antioxid Redox Signal 17: 794–802, 2012. doi:10.1089/ars.2011.4407.
    Crossref | PubMed | Web of Science | Google Scholar
  • 40. Panagopoulou P, Davos CH, Milner DJ, Varela E, Cameron J, Mann DL, Capetanaki Y. Desmin mediates TNF-α-induced aggregate formation and intercalated disk reorganization in heart failure. J Cell Biol 181: 761–775, 2008. doi:10.1083/jcb.200710049.
    Crossref | PubMed | Web of Science | Google Scholar
  • 41. Papp Z, van der Velden J, Stienen GJ. Calpain-I induced alterations in the cytoskeletal structure and impaired mechanical properties of single myocytes of rat heart. Cardiovasc Res 45: 981–993, 2000. doi:10.1016/S0008-6363(99)00374-0.
    Crossref | PubMed | Web of Science | Google Scholar
  • 42. Pattison JS, Osinska H, Robbins J. Atg7 induces basal autophagy and rescues autophagic deficiency in CryABR120G cardiomyocytes. Circ Res 109: 151–160, 2011. doi:10.1161/CIRCRESAHA.110.237339.
    Crossref | PubMed | Web of Science | Google Scholar
  • 43. Pyle WG, Solaro RJ. At the crossroads of myocardial signaling: the role of Z-discs in intracellular signaling and cardiac function. Circ Res 94: 296–305, 2004. doi:10.1161/01.RES.0000116143.74830.A9.
    Crossref | PubMed | Web of Science | Google Scholar
  • 44. Quintana E, Suri RM, Thalji NM, Daly RC, Dearani JA, Burkhart HM, Li Z, Enriquez-Sarano M, Schaff HV. Left ventricular dysfunction after mitral valve repair—the fallacy of “normal” preoperative myocardial function. J Thorac Cardiovasc Surg 148: 2752–2760, 2014. doi:10.1016/j.jtcvs.2014.07.029.
    Crossref | PubMed | Web of Science | Google Scholar
  • 45. Ryan TD, Rothstein EC, Aban I, Tallaj JA, Husain A, Lucchesi PA, Dell’Italia LJ. Left ventricular eccentric remodeling and matrix loss are mediated by bradykinin and precede cardiomyocyte elongation in rats with volume overload. J Am Coll Cardiol 49: 811–821, 2007. doi:10.1016/j.jacc.2006.06.083.
    Crossref | PubMed | Web of Science | Google Scholar
  • 46. Sanbe A, Osinska H, Saffitz JE, Glabe CG, Kayed R, Maloyan A, Robbins J. Desmin-related cardiomyopathy in transgenic mice: a cardiac amyloidosis. Proc Natl Acad Sci USA 101: 10132–10136, 2004. doi:10.1073/pnas.0401900101.
    Crossref | PubMed | Web of Science | Google Scholar
  • 47. Sanbe A, Yamauchi J, Miyamoto Y, Fujiwara Y, Murabe M, Tanoue A. Interruption of CryAB-amyloid oligomer formation by HSP22. J Biol Chem 282: 555–563, 2007. doi:10.1074/jbc.M605481200.
    Crossref | PubMed | Web of Science | Google Scholar
  • 48. Schiros CG, Dell’Italia LJ, Gladden JD, Clark D III, Aban I, Gupta H, Lloyd SG, McGiffin DC, Perry G, Denney TS Jr, Ahmed MI. Magnetic resonance imaging with 3-dimensional analysis of left ventricular remodeling in isolated mitral regurgitation: implications beyond dimensions. Circulation 125: 2334–2342, 2012. doi:10.1161/CIRCULATIONAHA.111.073239.
    Crossref | PubMed | Web of Science | Google Scholar
  • 49. Snider NT, Omary MB. Post-translational modifications of intermediate filament proteins: mechanisms and functions. Nat Rev Mol Cell Biol 15: 163–177, 2014. doi:10.1038/nrm3753.
    Crossref | PubMed | Web of Science | Google Scholar
  • 50. Ulasova E, Gladden JD, Chen Y, Zheng J, Pat B, Bradley W, Powell P, Zmijewski JW, Zelickson BR, Ballinger SW, Darley-Usmar V, Dell’Italia LJ. Loss of interstitial collagen causes structural and functional alterations of cardiomyocyte subsarcolemmal mitochondria in acute volume overload. J Mol Cell Cardiol 50: 147–156, 2011. doi:10.1016/j.yjmcc.2010.10.034.
    Crossref | PubMed | Web of Science | Google Scholar
  • 51. Wang X, Osinska H, Dorn GW II, Nieman M, Lorenz JN, Gerdes AM, Witt S, Kimball T, Gulick J, Robbins J. Mouse model of desmin-related cardiomyopathy. Circulation 103: 2402–2407, 2001. doi:10.1161/01.CIR.103.19.2402.
    Crossref | PubMed | Web of Science | Google Scholar
  • 52. Wang X, Osinska H, Klevitsky R, Gerdes AM, Nieman M, Lorenz J, Hewett T, Robbins J. Expression of R120G-αB-crystallin causes aberrant desmin and αB-crystallin aggregation and cardiomyopathy in mice. Circ Res 89: 84–91, 2001. doi:10.1161/hh1301.092688.
    Crossref | PubMed | Web of Science | Google Scholar
  • 53. Wang X, Sentex E, Saini HK, Chapman D, Dhalla NS. Upregulation of β-adrenergic receptors in heart failure due to volume overload. Am J Physiol Heart Circ Physiol 289: H151–H159, 2005. doi:10.1152/ajpheart.00066.2005.
    Link | Web of Science | Google Scholar
  • 54. Wei CC, Chen Y, Powell LC, Zheng J, Shi K, Bradley WE, Powell PC, Ahmad S, Ferrario CM, Dell’Italia LJ. Cardiac kallikrein-kinin system is upregulated in chronic volume overload and mediates an inflammatory induced collagen loss. PLoS One 7: e40110, 2012. doi:10.1371/journal.pone.0040110.
    Crossref | PubMed | Web of Science | Google Scholar
  • 55. Winter DL, Paulin D, Mericskay M, Li Z. Posttranslational modifications of desmin and their implication in biological processes and pathologies. Histochem Cell Biol 141: 1–16, 2014. doi:10.1007/s00418-013-1148-z.
    Crossref | PubMed | Web of Science | Google Scholar
  • 56. Yancey DM, Guichard JL, Ahmed MI, Zhou L, Murphy MP, Johnson MS, Benavides GA, Collawn J, Darley-Usmar V, Dell’Italia LJ. Cardiomyocyte mitochondrial oxidative stress and cytoskeletal breakdown in the heart with a primary volume overload. Am J Physiol Heart Circ Physiol 308: H651–H663, 2015. doi:10.1152/ajpheart.00638.2014.
    Link | Web of Science | Google Scholar
  • 57. Zheng J, Chen Y, Pat B, Dell’Italia LA, Tillson M, Dillon AR, Powell PC, Shi K, Shah N, Denney T, Husain A, Dell’Italia LJ. Microarray identifies extensive downregulation of noncollagen extracellular matrix and profibrotic growth factor genes in chronic isolated mitral regurgitation in the dog. Circulation 119: 2086–2095, 2009. doi:10.1161/CIRCULATIONAHA.108.826230.
    Crossref | PubMed | Web of Science | Google Scholar
  • 58. Zheng J, Yancey DM, Ahmed MI, Wei CC, Powell PC, Shanmugam M, Gupta H, Lloyd SG, McGiffin DC, Schiros CG, Denney TS Jr, Babu GJ, Dell’Italia LJ. Increased sarcolipin expression and adrenergic drive in humans with preserved left ventricular ejection fraction and chronic isolated mitral regurgitation. Circ Heart Fail 7: 194–202, 2014. doi:10.1161/CIRCHEARTFAILURE.113.000519.
    Crossref | PubMed | Web of Science | Google Scholar
  • 59. Zheng Q, Su H, Ranek MJ, Wang X. Autophagy and p62 in cardiac proteinopathy. Circ Res 109: 296–308, 2011. doi:10.1161/CIRCRESAHA.111.244707.
    Crossref | PubMed | Web of Science | Google Scholar

AUTHOR NOTES

  • *J. L. Guichard and M. Rogowski contributed equally to this work.

  • Address for reprint requests and other correspondence: L. J. Dell’Italia, Birmingham VA Medical Center, 700 S. 19th St., Birmingham, AL 35294-2180 (e-mail: ).