cells

Article Loss of Kinase Csnk2b/CK2β at Neuromuscular Junctions Affects Morphology and Dynamics of Aggregated Nicotinic Acetylcholine Receptors, Neuromuscular Transmission, and Synaptic Expression

Nane Eiber 1, Michael Rehman 1,2, Bojana Kravic 1,3, Rüdiger Rudolf 4 , Marco Sandri 5 and Said Hashemolhosseini 1,* 1 Institute of Biochemistry, Medical Faculty, Friedrich-Alexander-University of Erlangen-Nürnberg, 91054 Erlangen, Germany 2 Weill Cornell Medical College, Department of Medicine, New York, NY 10065, USA 3 Faculty of Biology, University of Duisburg-Essen, 45141 Essen, Germany 4 Institute of Molecular- and Cellular Biology, University of Applied Sciences Mannheim, 68163 Mannheim, Germany 5 Department of Biomedical Science, University of Padova, 35122 Padova, Italy * Correspondence: [email protected]; Tel.: +49–913–1852–4634

 Received: 28 June 2019; Accepted: 12 August 2019; Published: 20 August 2019 

Abstract: The Csnk2/CK2 is important for proliferation, differentiation, and survival. Previously, we showed that CK2 binds distinctive at neuromuscular junctions (NMJs) of mice and phosphorylates some of them. CK2 likely stabilizes clustered nicotinic acetylcholine receptors (AChRs). In the absence of the β-subunit of CK2 in skeletal muscle fibers, mice develop an age-dependent decrease of grip strength accompanied by NMJ fragmentation and impairments of neuromuscular transmission. However, the precise role of CK2β regarding the clustering of AChRs and downstream signaling at NMJs is unknown. Here, we compared conditional CK2β-deficient mice with controls and found in the mutants (1) a lower decrement of endplate potentials after repetitive stimulation and decrements of nerve-evoked compound muscle action potentials decayed more rapidly after synaptic transmission was partially blocked, (2) that their muscle weakness was partially rescued by administration of an acetylcholine esterase inhibitor, (3) fragmented NMJs and impaired AChR clustering was detected in muscles and cultured muscle cells, (4) enlarged myonuclei, (5) impaired synaptic , and (6) a high turnover rate of their AChR clusters in vivo. Altogether, our data demonstrate a role for CK2 at the NMJ by maintaining a high density of AChRs and ensuring physiological synaptic gene expression.

Keywords: Csnk2b; ; acetylcholine receptor; neuromuscular junction

1. Introduction The neuromuscular junction (NMJ) is a prototypical large synapse and the point of contact between motor nerve endings and a specialized surface area of muscle fibers [1]. Pre- and postsynaptic signaling pathways are mandatory for the formation and the maintenance of NMJs, and neuromuscular transmission is needed for muscle contraction. Motor nerve derived isoforms of a large heparan sulfate proteoglycoprotein called agrin are mandatory for NMJ formation and initiate a signaling cascade through interaction with their receptor Lrp4 on the surface of muscle fibers [2–4]. The receptor tyrosine kinase MuSK, part of a co-receptor with Lrp4, then becomes active and ensures both clustering of

Cells 2019, 8, 940; doi:10.3390/cells8080940 www.mdpi.com/journal/cells Cells 2019, 8, 940 2 of 16 nicotinic acetylcholine receptors (AChRs) and postsynaptic gene expression by a small number of myonuclei positioned underneath the postsynaptic membrane [2–6]. A pretzel-like shape reflects the morphology of clustered AChRs of adult muscle fibers, and these pretzels serve as hallmarks for physiological NMJs. Many perturbations of NMJs lead to structural disorders and are often represented by a fragmentation of the NMJ [1]. Several intracellular MuSK binding proteins have been discovered in recent decades, helping us to understand the signals necessary to build and maintain NMJs; some examples are cited here [7–11]. Previously, the protein kinase CK2 was identified by in vitro assays interacting with Rapsyn, Rac1, MuSK, Dok-7, and 14-3-3γ and phosphorylating the latter three [8,12]. The protein kinase Csnk2/CK2 is a tetramer composed of two catalytically active α- and two β-subunits; the α- subunits might be replaced by α’- subunits [13]. It phosphorylates and residues of target proteins and was reported as being important for cell proliferation, differentiation, and survival [13]. Although CK2 is expressed ubiquitously, accumulation in different cell compartments was reported [13]. A huge number of CK2 targets have been published and, as expected, constitutive CK2α or CK2β knockout mice are embryonically lethal [14,15]. A few years ago, CK2 was identified as interacting with MuSK and phosphorylating it [8]. Importantly, in the absence of CK2β in their muscle fibers, the mice lost grip strength in an age-dependent fashion. This muscle weakness was associated with heavily fragmented NMJs and impaired neuromuscular transmission. Later, MuSK was shown to also interact with Rapsyn, Rac1, Dok-7, and 14-3-3γ at NMJs and to phosphorylate the latter two [12]. Interestingly, phosphorylation of Rapsyn was very likely in silico but not experimentally confirmed [12]. Recently, it turned out that CK2 is also extra-synaptically important in muscle fibers, and its absence is associated with a compromised oxidative , consequently targeting mainly oxidative muscle fibers [16,17]. In fact, biopsies from human patients with mitochondrial myopathies are frequently associated with different transcript or activity profiles of CK2 [18]. Mechanistically, CK2 phosphorylates Tom22, the receptor of the mitochondrial protein import machinery, and thereby ensures muscle fiber homeostasis [16]. Here, we wanted to characterize the extent of the contribution of CK2 to the structure and the function of NMJs. We carefully investigated the muscle phenotype of conditional mutant mice, which do not express CK2β in their muscle fibers, in comparison with controls. Conditional knockout of CK2β in skeletal muscles of mice was ensured by human skeletal actin (HSA) driven Cre recombinase [8]. By blocking neuromuscular transmission in a dose-dependent manner, we unraveled the importance of CK2 activity for physiological neuromuscular transmission accompanied by significant changes of postsynaptic gene expression and a high turnover rate of AChR clusters.

2. Materials and Methods

2.1. Mice Strains Mouse experiments were performed in accordance with animal welfare laws and approved by the responsible local committees [animal protection officer, Sachgebiet Tierschutzangelegenheiten, FAU Erlangen-Nürnberg, AZ: I/39/EE006, TS-07/11, and Italian Ministry of Health, Office 5 (authorization number 1060/2015 PR)]. Mice were housed in cages that were maintained in a room with temperature 22 1 C and relative humidity 50–60% on a 12 h light/dark cycle. Water and food ± ◦ were provided ad libitum. Conditional CK2ß mice were described before [8]. Mice were genotyped by PCR analysis of tail biopsy DNA [8].

2.2. Grid Test, Treadmill Performance For the grip strength test, 8-month-old mice were put on the grid (made of 12 mm squares of 1 mm diameter wire), and the grid was turned upside down. The grid was kept in that position 40 cm above the ground for a period of 120 sec per repetition. There were five consecutive repetitions in total without resting time between repetitions. Neostigmine administration to mice was described Cells 2019, 8, 940 3 of 16 before [19]. Further, muscle force was measured with all four limbs by a Grip Strength Test Meter (Bioseb, Chaville, France). For assessment of physical endurance, a test based on treadmill performance was performed [20]. Mice were initially familiarized with the exercise protocol by walking or running on a rodent treadmill on consecutive days, during which they performed 20 min of exercise with 0% grade. A day later, mice were placed into a motorized wheel and allowed to run with a starting speed of 10 m per sec. After 10 min of exercise, the speed was increased by 2 m per sec each minute until the animals were exhausted. The experiment was repeated after 2, 4, and 24 h.

2.3. Nerve-Muscle Preparations and Extracellular Recordings Diaphragm-phrenic nerve preparations were maintained ex vivo in Liley’s solution gassed with 95% O2, 5% CO2 at room temperature [21]. The recording chamber had a volume of approximately 1 mL and was perfused at a rate of 1 mL/min. The nerve was drawn up into a suction electrode for stimulation with pulses of 0.1 ms duration. The preparation was placed on the stage of a Zeiss Axio Examiner Z1 microscope (Carl Zeiss MicroImaging, Göttingen, Germany) fitted with incident light fluorescence illumination with filters for 547 nm/red (Zeiss filter set 20) fluorescing fluorophore (Carl Zeiss MicroImaging). At the beginning of the experiment, the compound muscle action potential (CMAP) was recorded using a micropipette with a tip diameter of approximately 10 µm filled with bathing solution. The electrode was positioned so that the latency of the major negative peak was minimized. The electrode was then positioned 100 µm above the surface of the muscle, and CMAP was recorded. For recordings in the presence of tubocurarine, the chamber was filled with 2 mL (300 nM, 500 nM, 800 nM, or 1000 nM) of d-tubocurarine chloride (Sigma Aldrich Chemie, München, Germany). During the curare treatment, trains of 25 repetitive nerve stimulations (5 Hz) were performed at 2 min intervals, and the ratio of CMAP amplitudes (mean (20th–25th)/2nd) was calculated [22].

2.4. Intracellular Recordings and Data Analysis To block muscle action potentials so that EPPs (endplate potentials) and EPCs (endplate currents) could be recorded [23,24], µ-conotoxin GIIIB (µ-CTX, 2 µM; Peptide Institute, Osaka, Japan) was added to Lilly’s solution. Concurrently, clustered AChRs at NMJs were labeled by adding 0.5 10 8 M × − of rhodamine-α-bungarotoxin (Life Technologies, Darmstadt, Germany) to the same Lilly solution. In some experiments, the effect of the toxin wore off after 1–2 h, and contractions resumed in response to nerve stimulation. These preparations were then exposed a second time to the toxin. Two intracellular electrodes (resistance 10–15 MΩ) were inserted within 50 µm of the NMJs under visual inspection [24]. Current was passed through one electrode to maintain the membrane potential within 2 mV of 75 mV, − while voltage transients were recorded with the other. Signals were amplified by an Axoclamp 900 A and digitized at 40 kHz by a Digidata 1440 A under the control of pCLAMP 10 (Molecular Devices, Sunny Vale, CA, USA). Voltage records were filtered at 3 kHz and current records at 1 kHz (8-pole Bessel filter). Current transients were recorded using the two-electrode voltage-clamp facility of the Axoclamp 900 A. Clamp gains were usually 300–1000, reducing the voltage transients to <3% of their unclamped amplitudes. At most NMJs, 50–100 spontaneous quantal events were recorded during a period of 1 min. Records were analyzed using pCLAMP 10. Spontaneous events were extracted using the “template search” facility and edited by eye to remove obvious artifacts. Events recorded from each NMJ were averaged, and the amplitude and frequency were determined.

2.5. Quantitative 3D Morphometrical Imaging

Mice were killed by CO2 affixation. Mouse soleus muscle was dissected and fixed in 2% paraformaldehyde (PFA) for 2 h at 4 ◦C. Muscle bundles containing 5–10 fibers were prepared and stained with bungarotoxin (BTX) 1:2.500 (Rh-BTX, Invitrogen, Darmstadt, Germany) for 1 h at room temperature. Stained bundles were washed three times 5 min in phosphate buffered saline (PBS) and embedded in Mowiol. Then, 3D images of NMJs were taken with a 40 oil objective (Zeiss Examiner × Cells 2019, 8, 940 4 of 16

E1, Carl Zeiss MicroImaging) at 55 ms exposure time. Images were deconvoluted and analyzed using different modules in AxioVision Software (ZEISS AxioVision Release 4.8, Carl Zeiss MicroImaging)). The following parameters were determined for each NMJ: volume, surface, grey sum, grey mean, number of fragments. For each genotype, more than 50 NMJs were analyzed.

2.6. Tissue Sections, Immunohistochemistry For immunofluorescence analysis, all muscles were quick-frozen in prechilled isopentane and embedded in Tissue-Tec (Leica Instruments, Wetzlar, Germany). Muscles were cryotome-sectioned to 10 µm slices. Unfixed sections were rinsed with PBS and permeabilized for 5–10 min in PBS supplemented with 0.1% Triton X-100, then blocked in 10% fetal calf serum (FCS) and 1% bovine serum albumin (BSA) for 1–2 h. Cryotome sections were used for immunofluorescence stainings. For immunofluorescent stainings, sections were washed for 5 min in PBS, permeabilized in 0.1% Triton X-100 for 10 min, and blocked in PBS solution containing 10% FCS and 1% BSA for 1 h at 25 ◦C. Rhodamine- or Alexa 647 conjugated bungarotoxin 1:2.500 (Rh BTX; Thermo Fisher Scientific, Schwerte, Germany) was used for NMJ detection. Stained cryosections were analyzed and documented using a Zeiss Axio Examiner Z1 microscope (Carl Zeiss MicroImaging)) equipped with an AxioCam MRm camera (Carl Zeiss MicroImaging) and ZEISS AxioVision Release 4.8 (Carl Zeiss MicroImaging). Nuclei diameters were analyzed with the Scion Image Software (Scion Corporation, Frederick, MD, USA).

2.7. Tissue Culture, Culturing of Primary Muscle Cells, Transfection Primary skeletal muscle cells were prepared from leg muscles of neonatal mice. The tissue was minced with a razor blade and dissociated with collagenase type IV (Sigma Aldrich Chemie, Taufkirchen, Germany) and Dispase II (Roche Diagnostics, Mannheim, Germany) in Mg2+- and Ca2+-free Hank´s Balanced Salt Solution (HBSS) buffer. Digestion was stopped by adding FCS. After filtering through a cell strainer (Invitrogen) and centrifugation, cells were resuspended in 80% Ham’s F10 (Ca2+-free medium), 20% FCS, 1% penicillin/streptomycin, and recombinant human fibroblast growth factor (5 ng/mL) (Promega Corporation, Fitchburg, MA, USA). Subsequently, cells were seeded on Matrigel-coated plates (Invitrogen, Carlsbad, CA, USA). After 24 h, the culture medium was replaced by 40% Dulbecco’s Modified Eagle Medium (DMEM), 40% Ham’s F10, 20% FCS, 1% penicillin/streptomycin, and recombinant human fibroblast growth factor (5 ng/mL). As soon as primary skeletal muscle cells reached confluence, the medium was replaced for differentiation by 98% DMEM, 2% horse serum, and 1% penicillin/streptomycin. The production of agrin-conditioned media was described previously [25]. Agrin-conditioned medium was added at 1:8 dilution to primary myotubes. AChR aggregates were detected or quantified 16 h later, as described below. The terms active and inactive reflect agrin originating either from isoform agrinA0B0 or agrinA4B8 [26]. Quantification of different AChR endplate types was performed by analyzing >50 AChR aggregates for maximal length and surface area using ZEISS Axiovision software modules AutoMeasure Plus and Commander (Carl Zeiss MicroImaging).

2.8. DNA and RNA Preparation, Reverse Transcription, PCR Total RNA was extracted from mouse diaphragm, gastrocnemius, and soleus muscle with TRIzol reagent (Life Technologies, Darmstadt, Germany) [8]. After reverse transcription, cDNAs were used with mouse-specific primers (see Table1) for quantitative PCR reactions using the ABsolute QPCR SYBR Green Capillary Mix (Thermo Fisher Scientific, Schwerte, Germany) and the glass capillaries and Lightcycler Thermal Cycle System (Roche Diagnostics, Mannheim, Germany). Data analysis was performed as described elsewhere [8]. Cells 2019, 8, 940 5 of 16

Table 1. Oligonucleotides.

Gene Primer Sequence 5 -CTACCTGCCCACAGACTCAG-3 AChRα 0 0 50-TTGGACTCCTGGTCTGACTT-30 5 -GGTCAATGTCAGCCTGAAGC-3 AChRγ 0 0 50-GCACATGCATCCGTAACAGC-30 5 -TCTCCAACTATGATAGCTCGGT-3 AChRβ 0 0 50-CATTGATGTCCAGGGCAACGTC-30 5 -TGTATGGCTGCCAGAGATTG-3 AChRε 0 0 50-GCGGATGATGAGCGTATAGA-30 5 -ATGAGGAACAAAGGCTGATCCA-3 AChRδ 0 0 50-ACAGTGATGTTCCCGAAGTCGT-30 5 -CCGCTACAGGCACTCTGTCT-3 Rapsyn 0 0 50-TCAGTCTCCTCCACGCACTC-30 5 -GCCTTGGTTGAAGAAGTAGC -3 MuSK 0 0 50-CTTGATCCAGGACACAGATG -30 5 -GTTCGTGTACTGCGGCAAGA-3 RPL8 0 0 50-ACAGGATTCATGGCCACACC-30

2.9. In Vivo Microscopy and Analysis of AChR Turnover and NMJ Fragmentation In vivo microscopy of mice was performed under anesthesia using Zoletil and xylazine on a Leica SP2 confocal microscope (Leica Instruments, Wetzlar, Germany) equipped with a 63 1.2 numerical × aperture water immersion objective, essentially as described previously [27,28]. Automated analysis of AChR turnover and NMJ fragmentation used algorithms described earlier [28]. Quantification of mean intensities of BTX-AF647 or BTX-AF555 labeled NMJs were done using ImageJ (NIH). To quantify the mean intensity, each NMJ was manually cropped out from a confocal stack. Then, using the tool “sum of slices”, a projection of the stack was created. In total, >15 NMJs were analyzed of both control and CK2β-deficient muscles.

2.10. Statistical Analysis Data are presented as the mean values, and the error bars indicate s.e.m. The significance was ± calculated by unpaired two-tailed t test or as indicated by the figure legends and is provided as real p-values believed to be categorized for different significance levels, such as **** p < 0.0001, *** p < 0.001, ** p < 0.01, or * p < 0.05.

3. Results

3.1. The Ablation of CKβ in Skeletal Muscle Fibers of Mice Results in Impaired Neuromuscular Transmission and Reduced Grip Strength Previously, we recorded a difference of miniature endplate current amplitudes by comparing diaphragm muscle fibers of conditional CK2β-deficient and control mice, especially opposing control muscle fibers with regular NMJs with mutant fibers and structurally fragmented NMJs [8,16]. In order to better understand the properties of neuromuscular transmission in the CK2β-deficient mice, we measured a number of electrophysiological parameters in CK2β-deficient mutant diaphragm fibers with fragmented NMJs and control muscle fibers with unaffected NMJs. Muscle fiber action potentials were blocked by µ-conotoxin GIIIB (µCTX, 2–3 µM) to prevent contraction, as described before [29]. For these studies, we used either 3-months-old or >6-months-old mice. Next to membrane resistance, miniature endplate potentials and nerve evoked endplate potentials as well as time course and amplitudes of currents were recorded using a two-electrode voltage clamp. Although the muscles were exposed to rhodamine fluorophore coupled bungarotoxin (BTX) using a concentration of 10 nM Cells 2019, 8, 940 6 of 16 for 30–40 min, this did not cause any significant diminution of miniature endplate potential (mEPP) amplitude (~0.8 mV, Figure1B) compared to previously reported values [ 30]. We did not detect a change of membrane resistance between CK2β-deficient and control mice in either 3-months-old or in >6-months-old mice (Figure1A). Amplitudes of nerve-independent miniature endplate potentials and currents were significantly changed in >6-months-old CK2β-deficient mice in comparison to control litters but not in 3-months-old mice (Figure1B,C). Noteworthy, we did not detect any change in mEPP frequencyCells 2019, 8, in x control and CK2β-deficient diaphragm muscle fibers (Figure6 1ofD). 17 Recording of nerve-evokednot in endplate3-months-old potentials mice (Figure 1B,C). and currentsNoteworthy, did we did not not reveal detect any any change changes in mEPP between frequency control and CK2β-deficientin control diaphragm and CK2β muscle-deficient fibersdiaphragm (Figure muscle1E,F). fibers Our(Figure first 1D). step Recording in identifying of nerve-evoked nerve-evoked endplate potentials and currents did not reveal any changes between control and CK2β β-deficient impairmentsdiaphragm failed, as muscle we did fibers not (Figure detect 1E,F). any Our change first step betweenin identifying control nerve-evoked and CK2impairments-deficient failed, diaphragm muscle fibersas while we did comparingnot detect any ch theange decrement between control of and endplate CK2β-deficient potential diaphragm amplitude muscle fibers at 5 while Hz (Figure1G). In this case, thecomparing diaphragm the decrement muscle of wasendpla stimulatedte potential amplitude by 25 sweeps at 5 Hz within(Figure 1G). approximately In this case, the 3–4 s. diaphragm muscle was stimulated by 25 sweeps within approximately 3–4 s.

Figure 1. In >Figure6-months-old 1. In >6-months-old mice but mice not 3-months-oldbut not 3-months-old mice, mice, conditional conditional CK2 CK2ββ-deficient-deficient mice mice exhibited exhibited impairment of neural transmission, as reflected by lowered miniature endplate potentials impairment of neural transmission, as reflected by lowered miniature endplate potentials and currents and currents in comparison with wild-type litters. The following electrophysiological recordings are in comparisonpresented: with wild-type(A) membrane litters. resistance, The (B following) miniature endplate electrophysiological potential amplitudes, recordings (C) miniature are presented: (A) membraneendplate resistance, current ( Bamplitudes,) miniature (D endplate) frequency potential of miniature amplitudes, endplates, ((EC) )endplate miniature potential endplate current amplitudes, (F) endplate current amplitudes, (G) decrement of endplate potential amplitude at 5 Hz. amplitudes, (D) frequency of miniature endplates, (E) endplate potential amplitudes, (F) endplate (*** p < 0.001; ** p < 0.01; unpaired two-tailed Student’s t-test; mice per genotype ≥ 3). current amplitudes, (G) decrement of endplate potential amplitude at 5 Hz. (*** p < 0.001; ** p < 0.01; unpaired two-tailedWe wanted Student’s to understand t-test; micethe extent per genotype of muscle weakness3). in response to compromised neuromuscular transmission in more detail. We conducted≥ several behavioral studies comparing We wantedcontrol toandunderstand conditional CK2 theβ-deficient extent mice. of Compared muscle with weakness control littermates, in response conditional to mice compromised that lacked the β-subunit of CK2 in skeletal muscle fibers had a loss of grip strength that started by neuromuscularapproximately transmission three months in more of age detail. and did We not conductedfurther progress several after an behavioral additional 6–12 studies weeks comparing control and conditional(Figure 2A,B). Loss CK2 ofβ grip-deficient strength was mice. measured Compared by either witha grip-strength control meter littermates, (Figure 2A) conditional or by mice that lacked thecountingβ-subunit the time mice of CK2 were able in skeletalto cling upside muscle down fiberson a grid had (Figure a 2B,C); loss ofgrip grip strength strength loss was that started even more prominent after repetitive upside-down clinging on a grid (Figure 2A,B). Further, the grip by approximatelystrength three of mutant months mice ofcould age only and be did partly not rescued further by progress intravenous after administration an additional of the 6–12 weeks (Figure2A,B).acetylcholine Loss of grip esterase strength inhibitor was neostigmine measured (Figure by 2C). either In accordance a grip-strength with these data, meter conditional (Figure 2A) or by counting theCK2 timeβ-deficient mice weremice fatigued able to earlier cling during upside running down wheel on endurance a grid training (Figure in 2comparisonB,C); grip with strength loss control mice (Figure 2D). Overall, these data demonstrate a compromised neuromuscular was even moretransmission prominent but do afternot disclose repetitive how nerve-evoked upside-down muscle weakness clinging occurs. on a grid (Figure2A,B). Further, the grip strength of mutant mice could only be partly rescued by intravenous administration of the acetylcholine esterase inhibitor neostigmine (Figure2C). In accordance with these data, conditional CK2β-deficient mice fatigued earlier during running wheel endurance training in comparison with control mice (Figure2D). Overall, these data demonstrate a compromised neuromuscular transmission but do not disclose how nerve-evoked muscle weakness occurs. Cells 2019, 8, 940 7 of 16

Cells 2019, 8, x 7 of 17

Figure 2. GripFigure strength 2. Grip of strength muscle-specific of muscle-specific CK2 CK2β-deficientβ-deficient mice. mice. (A (A) Grip) Grip strength strength of >5 different of >5 di pairsfferent pairs of mice (each mouse pair was composed of wild-type and mutant litters) of indicated age was of mice (eachmeasured mouse pair by a was grip-strength composed meter of wild-type(Bio-GS3 Grip-Test, and mutant Bioseb litters) Vitrolles, of indicatedFrance). (*** agep < was0.001; measured by a grip-strengthunpaired meter two-tailed (Bio-GS3 Student’s Grip-Test, t-test; n = Bioseb >5 mice, Vitrolles, each genotype). France). Error (***barsp indicate< 0.001; s.e.m unpaired (B) Muscle two-tailed Student’s t-test;gripn strength= >5 mice,was determined each genotype). five consecutive Error times bars by indicatemeasurings.e.m the time (B mice) Muscle were able grip to strengthcling was determined fiveupside-down consecutive on a timesgrid until by they measuring fell down. the Mice time were mice then wereimmediately able toput cling back upside-downon the grid, and on a grid the drop time was measured again. The first maximal clinging time measurement was set to 100%. until they fellThe down. next Micefour maximal were then clinging immediately times decreased put co backntinuously. on the Different grid, and colors the symbolize drop time different was measured again. The firstmouse maximal ages of clingingCK2β-deficient time mice. measurement As indicated, was 140 up set to to 480 100%. days old The mice next were four analyzed. maximal (C) clinging times decreasedMuscle continuously. grip strength Di wasfferent measured colors after symbolize administration different of mouseacetylcho agesline esterase of CK2 βinhibitor-deficient mice. As indicated,(neostigmine) 140 up to 480to CK2 daysβ-deficient old mice mice were(tail vein analyzed. injection). After (C) Muscleindicated griptimes, strength maximal upside- was measured down clinging time was measured and plotted to the number of repetitions. Note that wild-type mice after administrationwere able ofto acetylcholinecling up to 10 min esterase upside-down inhibitor on a grid, (neostigmine) while mutant mice to CK2after neostigmineβ-deficient injection mice (tail vein injection). Afterwere indicatedonly able to times,cling upside-down maximal for upside-down about 50 s. (D) clingingEndurance time capacity was of measuredaged-matched and wild- plotted to the number oftype repetitions. and mutant mice Note (8–10 that months wild-type of age) was mice determined were by able enforced to cling runni upng on to a 10treadmill. min upside-down Note that mutant mice were exhausted at lower treadmill speed. on a grid, while mutant mice after neostigmine injection were only able to cling upside-down for about 503.2. s. ( DIn) the Endurance Absence of the capacity CK2β Subunit of aged-matched the Function of wild-type the NMJ is andImpaired mutant mice (8–10 months of age) was determinedTo better by enforced understand running muscle on fatigability, a treadmill. reduced Note thatnerve-independent mutant mice werechanges exhausted of miniature at lower treadmillendplate speed. potentials, and currents in conditional CK2β-deficient mice in comparison with controls, further electrophysiological recordings were performed with phrenic nerve-diaphragm explants of 3.2. In the Absencewild-type of theand CK2 mutantβ Subunit mice. The the reduced Function mEPP/miniature of the NMJ Isendplate Impaired current (mEPC) amplitudes could have been due to either decreased quantal size or to a reduced postsynaptic sensitivity to To betterindividual understand quanta.muscle Notably, fatigability,both above-mentioned reduced neuromuscular nerve-independent transmission changes defects were of miniature endplate potentials,observed, andas the currents mean quanta in conditional were significantly CK2 βreduced-deficient in CK2 miceβ-deficient in comparison muscles (data with not controls, further electrophysiologicalshown). We next assessed recordings neuromuscular were transmission performed by with repetitive phrenic stimulat nerve-diaphragmion of the phrenic nerve explants of with 20 Hz trains for 10 s and calculated the decrement of EPP (Figure 3A,B). We did not measure wild-type and mutant mice. The reduced mEPP/miniature endplate current (mEPC) amplitudes could any decrease of decrement in 3-months-old mice (Figure 3A), but there was a significant decrease of have been duedecrement to either starting decreased after 5 s in quantal >6-months-old size or mice to (F aigure reduced 3B). We postsynaptic finally evaluated sensitivity the safety factor, to individual quanta. Notably,which bothreflected above-mentioned the fact that the threshold neuromuscular required to transmission generate a muscle defects action were potential observed, was as the mean quantaexceeded were significantly by the excitatory reduced effect generated in CK2β by-deficient nerve stimulation muscles [31]. (data Towards not shown).this aim, we We carried next assessed out compound muscle action potential (CMAP) measurements on wild-type and CK2β-deficient neuromusculardiaphragms transmission in the presence by repetitive of increasing stimulation concentrations of the of D phrenic-tubocurarine nerve in order with to 20 monitor Hz trains the for 10 s and calculated the decrement of EPP (Figure3A,B). We did not measure any decrease of decrement in 3-months-old mice (Figure3A), but there was a significant decrease of decrement starting after 5 s in >6-months-old mice (Figure3B). We finally evaluated the safety factor, which reflected the fact that the threshold required to generate a muscle action potential was exceeded by the excitatory effect generated by nerve stimulation [31]. Towards this aim, we carried out compound muscle action potential (CMAP) measurements on wild-type and CK2β-deficient diaphragms in the presence of increasing concentrations of d-tubocurarine in order to monitor the effect of a partial block of AChRs. Treatment with d-tubocurarine led to a dose-dependent strong decrease of the decrement in response to repetitive stimuli in CK2β-deficient muscles but not in wild-type muscles in >6-months-old mice Cells 2019, 8, 940 8 of 16

(Figure3C,D), pointing at a reduced safety factor in CK2 β-deficient mice. Obviously, a concentration range of 300–800 nM tubocurarine resulted in a stronger decrease of the decrement in mutant muscles, while high concentrations of tubocurarine ( 1000 nM) were presumably blocking too many AChRs, ≥ making it impossible to detect a change between controls and mutants (Figure3D). Importantly, diaphragms of 3-months-old mice were similarly sensitive to tubocurarine regardless of their genotype (Figure3C). These results indicate that lack of CK2 β in muscle fibers impairs neuromuscular synaptic transmission by affecting both sustained neurotransmitter release and safety factor. Cells 2019, 8, x 9 of 17

Figure 3. CK2β-ablation in skeletal muscles altered neuromuscular synapse function and affected Figure 3. CK2β-ablation in skeletal muscles altered neuromuscular synapse function and affected safety safety factor in >6-months-old but not in 3-months-old conditional CK2β deficient mice. (A,B) factor in >6-months-oldDecrement of the but endplate not in potential 3-months-old amplitude conditional in wild-type and CK2 CK2ββ-deficientdeficient diaphragm mice. ( Amuscles,B) Decrement of the endplate potentialin 3-months-old amplitude mice (A) and in wild-type>6-months-old and mice. CK2 (C,Dβ) -deficientCompound muscle diaphragm action potential muscles (CMAP) in 3-months-old mice (A) and measurements>6-months-old in 3-months-old mice. (C ,(DC) )and Compound >6-months-old muscle (D) wild-type action and potential CK2β-deficient (CMAP) diaphragms measurements in under untreated conditions and in the presence of increasing concentrations (300, 500, 800, and 1,000 3-months-oldnM) (C )of and D-tubocurarine.>6-months-old The >6-months-old (D) wild-type CK2β-deficient and CK2 animalsβ-deficient showed diaphragmsa significantly higher under untreated conditions anddecrement in the of CMAP, presence which of was increasing already displayed concentrations at 300 nM D-tubocurarine, (300, 500, thus 800, unmasking and1,000 a nM) of d-tubocurarine.reduced The safety>6-months-old factor in the absence CK2 βof-deficient CK2β (*** p < animals 0.001; ** p showed< 0.01; * p < a 0.05; significantly unpaired two-tailed higher decrement Student’s t-test; n = 3–5 mice, each genotype). Error bars indicate s.e.m. of CMAP, which was already displayed at 300 nM d-tubocurarine, thus unmasking a reduced safety

factor in the absence of CK2β (*** p < 0.001; ** p < 0.01; * p < 0.05; unpaired two-tailed Student’s t-test; n = 3–5 mice, each genotype). Error bars indicate s.e.m. CellsCells2019 2019, 8, ,8 940, x 109 ofof 1617

3.3. Structural Changes of Clustered AChRs in CK2β-Deficient Myofibers and Cultured Myotubes 3.3. Structural Changes of Clustered AChRs in CK2β-Deficient Myofibers and Cultured Myotubes To explore the in vivo role of CK2β at the NMJ, we performed whole-mount BTX stainings of adultTo >6-months-old explore the in wild-type vivo role ofand CK2 mutantβ at the mo NMJ,use muscles. we performed Postsynaptic whole-mount boutons BTXof CK2 stainingsβ muscles of adultappeared>6-months-old less regular wild-type in shape and and frequently mutant mouse fragmented muscles. (Figures Postsynaptic 4A and boutons 5A,B). Fragmentation of CK2β muscles was appearedconfirmed less by regularmorphometric in shape analysis and frequently of the soleus fragmented muscle, (Figures revealing4A anda marked5A,B). Fragmentationfragmentation of wasNMJs confirmed in CK2 byβ-deficient morphometric muscles analysis when ofcompared the soleus to muscle, wild-type revealing muscles a marked (Figure fragmentation 4A). In CK2 ofβ- NMJsdeficient in CK2 soleus,β-deficient more musclesthan half when of the compared NMJs were to wild-type assembled muscles from (Figure more 4thanA). In five CK2 fragmentsβ-deficient in soleus,comparison more with than halfcontrols of the (Figure NMJs 4A). were In assembled the soleus frommuscle, more NMJ than fragmentation five fragments was in accompanied comparison withby changes controls (Figurein 3D 4morphometricA). In the soleus parameters muscle, NMJ with fragmentation an increase was of accompaniedsurface area, by volume, changes and in 3Dfluorescence morphometric intensity parameters of NMJs with (Figures an increase 4B–E and of surface 5A,B). area,Endplate volume, fragmentation and fluorescence was often intensity due to ofaging NMJs or (Figuremuscle4 denervationB–E and Figure [32,33].5A,B). We Endplate asked whether fragmentation these changes was oftenof AChR due clustering to aging or might muscle have denervationbeen imitated [32 ,in33 cultured]. We asked cells. whether In accordance these changes with ofour AChR previously clustering published might havedata been[8], treatment imitated in of culturedprimary cells.cultured In accordance myotubes with from our CK2 previouslyβ-deficient published skeletal data muscles [8], treatment by agrin-conditioned of primary cultured media myotubesinduced formation from CK2 βof-deficient aggregated skeletal AChRs muscles of significantly by agrin-conditioned smaller size mediain comparison induced formationwith controls of aggregated(Figure 4F,G). AChRs of significantly smaller size in comparison with controls (Figure4F,G).

FigureFigure 4. 4.Neuromuscular Neuromuscular junctions junctions (NMJ) (NMJ) morphometric morphometric analysis analysis of of wild-type wild-type and and CK2 CK2ββ-deficient-deficient skeletalskeletal musclesmuscles and and myoblasts. myoblasts. ((AA)) MorphometricMorphometric analysisanalysis ofof whole-mountwhole-mount bungarotoxinbungarotoxin (BTX)(BTX) stainingstaining of of muscles muscles of of wildtype wildtype and and CK2 CK2ββ-deficient-deficient mice. mice. Mutant Mutant muscles muscles lacking lacking CK2 CK2ββdisplayed displayed a a significantlysignificantly higher higher NMJ NMJ fragmentation fragmentation when when compared compared to to wild-type wild-type muscles; muscles; (** (**p p< <0.01; 0.01; * *p p< <0.05; 0.05; unpairedunpaired two-tailed two-tailed Student’s Student’s t-test; t-test;n n> >50 50NMJs NMJsper per genotypegenotypefrom fromat at leastleast threethree micemice perper genotype).genotype). ErrorError bars bars indicate indicate s.e.m. s.e.m. (B–E ()B Morphometric–E) Morphometric analysis analysis of surface of areasurface (B), volumearea (B (C),), volume total fluorescence (C), total intensityfluorescence (D), andintensity mean (D fluorescence), and mean intensity fluorescence (E) of intensity NMJs in (E whole-mount) of NMJs in whole-mount preparations preparations of soleus of wild-typeof soleus andof wild-type conditional and CK2 conditionalβ-deficient CK2 miceβ-deficient following mice staining following with BTX. staining All thewith analyzed BTX. All NMJ the featuresanalyzed were NMJ significantly features alteredwere significantly in CK2β-deficient altered muscles in CK2 exceptβ-deficient for mean muscles fluorescence except intensity for mean (E) whenfluorescence compared intensity to wild-type (E) when muscles compared (*** p to< wild-type0.001, ** p muscles<0.01; *, (***p < p0.05; < 0.001, n.s. ** means p <0.01; not *,significant; p <0.05; n.s. unpairedmeans not two-tailed significant; Student’s unpaired t-test; two-tail soleus wild-type;ed Student’sn > t-test;50 NMJs, soleus CK2 wild-type;β-deficient n soleus > 50 nNMJs,> 50 NMJs). CK2β- NMJsdeficient were soleus sampled n > 50 from NMJs). three NMJs mice were per genotype. sampled from Error thr barsee mice indicate per genotype. s.e.m. (F, GError) Morphometric bars indicate analysiss.e.m. (F of, lengthG) Morphometric (F) and fluorescence analysis intensity of length per (F area) and (G )fluorescence was performed intensity with clusteredper area and(G) BTX was stainedperformed acetylcholine with clustered receptors and (AChRs) BTX stained on cultured acetylcholine primary receptors myotubes. (AChRs) Clusters on were cultured sampled primary from threemyotubes. primary Clusters muscle were cell extractssampled per from genotype. three primar Errory bars muscle indicate cell s.e.m.extracts per genotype. Error bars indicate s.e.m.

Cells 2019, 8, 940 10 of 16 Cells 2019, 8, x 11 of 17

Figure 5. Conspicuous morphological alterations were visible at the NMJs of CK2β-deficient skeletal Figure 5. Conspicuous morphological alterations were visible at the NMJs of CK2β-deficient skeletal muscles. (A, B) Representative 2D (A) and 3D (B) images of whole-mount stainings of wild-type and muscles. (A, B) Representative 2D (A) and 3D (B) images of whole-mount stainings of wild-type CK2β-deficient soleus muscle after BTX staining (red) showing the presence of irregular and and CK2β-deficient soleus muscle after BTX staining (red) showing the presence of irregular and fragmented NMJs in CK2β-deficient-deficient muscles. Scale Scale bar, bar, 20 µm.µm.

3.4. Changes Changes of of Postsynaptic Postsynaptic Gene Expression Unraveled in CK2 β-Deficient-Deficient Muscle Fibers We wondered whetherwhether thesethese structuralstructural changeschanges of the postsynaptic machinery in CK2β-deficient-deficient fibersfibers were were accompanied accompanied with with molecular molecular differences differences in inpostsynaptic postsynaptic gene gene transcription. transcription. It is Itknown is known that transcriptionalthat transcriptional activity activity might might change change the size the of size nuclei of nuclei[34]. To [34 gain]. To a first gain insight, a first insight, we analyzed we analyzed muscle cellmuscle nuclei cell diameters. nuclei diameters. By comparin By comparingg the cell thenuclei cell diameters nuclei diameters of CK2β of-deficient CK2β-deficient and control and controlmuscle fibers,muscle we fibers, detected we detecteda strong increase a strong in increase the size in of the nuclei size in of mutant nuclei infibers mutant (Figure fibers 6A). (Figure Next, 6weA). looked Next, weat transcript looked at levels transcript of AChR levels subunits of AChR by subunits comparing by comparingdiaphragm, diaphragm, soleus, and soleus,gastrocnemius and gastrocnemius muscles of CK2musclesβ-deficient of CK2 miceβ-deficient with controls mice with (Figure controls 6B,C). (Figure Importantly,6B,C). Importantly, mRNA levels mRNA of AChR levelsβ and of AChR AChRβδ subunitsand AChR wereδ subunits markedly were increased markedly in increasedmutant muscles, in mutant while muscles, AChRα while and AChRεα wereand AChRincreasedε were in someincreased of the in muscles some of (Figure the muscles 6B). We (Figure never6B). observed We never a decrease observed in a mRNA decrease levels in mRNA regarding levels AChR regarding AChRin CK2 genesβ-deficient in CK2 musclesβ-deficient (Figure muscles 6B,C). (Figure Moreover,6B,C). CK2 Moreover,β-deficient CK2 musclesβ-deficient displayed muscles a displayedremarkable a upregulationremarkable upregulation of AChRγ ofand AChR MuSK,γ and but MuSK, not Rapsyn but not Rapsynor mRNA or mRNAtranscripts transcripts (Figure (Figure 6C–E),6C–E), thus confirmingthus confirming NMJ alterations NMJ alterations and pointing and pointing to an abno tormal an abnormal neuromuscular neuromuscular transmission transmission [35]. In order [35 to]. betterIn order understand to better understand the influence the influenceof CK2 activity of CK2 activityon the expression on the expression of postsynaptic of postsynaptic genes, genes, we also we performedalso performed cell culture cell culture studies studies with two with different two diff commonerent common CK2 inhibitors, CK2 inhibitors, Apigenin Apigenin and DMAT, and DMAT, which whichwere used were successfully used successfully before beforewith cultured with cultured muscle muscle cells [8]. cells It turned [8]. It turnedout that out both that inhibitors both inhibitors had a negativehad a negative influence influence on the onexpression the expression of AChR ofα AChR, AChRαγ,, AChR MuSK,γ, and MuSK, Rapsyn; and Rapsyn;some of somethese changes of these werechanges evidenced were evidenced as being statisti as beingcally statistically significant significant (Figure 6F). (Figure 6F).

Cells 2019, 8, 940 11 of 16 Cells 2019, 8, x 12 of 17

FigureFigure 6. 6. GeneGene expressionexpression levels levels for for postsynaptic postsynaptic genes genes in the in absence the absence of CK2 ofβ .(CK2A) Analysisβ. (A) Analysis of cell of cell nucleinuclei diameter diameter inin soleussoleus muscle muscle of of wild-type wild-type and and conditional conditional CK2β CK2-deficientβ-deficient mice. Analyzedmice. Analyzed cell cell nucleinuclei were were significantlysignificantly bigger bigger in in diameter diameter in CK2in CK2β-deficientβ-deficient muscles muscles when when compared compared to wild-type to wild-type cellcell nuclei nuclei (**** pp << 0.0001;0.0001; unpaired unpaired two-tailed two-tailed Student’s Student’s t-test; t-test; soleus soleus wild-type; wild-type;n > 50 n cell > nuclei,50 cell nuclei, CK2CK2ββ-deficient-deficient soleus soleus nn >> 50 cell nuclei). CellCell nucleinuclei were were sampled sampled from from three three mice mice per per genotype. genotype. (B– (B–E) Relative transcript levels for AChRα, AChRβ, AChRδ, AChRε (B), AChRγ (C), MuSK (D), E) Relative transcript levels for AChRα, AChRβ, AChRδ, AChRε (B), AChRγ (C), MuSK (D), and and Rapsyn (E) in diaphragm, gastrocnemius, and soleus muscle, as determined by quantitative PCR. Rapsyn (E) in diaphragm, gastrocnemius, and soleus muscle, as determined by quantitative PCR. Relative transcript levels of AChRβ, AChRδ (B), AChRγ (C), and MuSK (D) were significantly elevated Relativein diaphragm, transcript soleus, levels and extensor of AChR digitorumβ, AChR longusδ (B (EDL)), AChR (***pγ< 0.001,(C), and ** p 3 musclesand extensor per genotype). digitorum (F) longus After incubation (EDL) (*** ofp C2C12 < 0.001, muscle ** p cells< 0.01; * p < 0.05;with unpaired common CK2two-tailed inhibitors Student’s Apigenin t-test; and DMAT, n > 3 muscles the transcript per genotype). levels of typical (F) After postsynaptic incubation genes of C2C12 musclewere significantly cells with reducedcommon (**** CK2p < inhibitors0.0001, *** pApigenin< 0.001, ** andp < 0.01;DMAT, * p < the0.05; transcript unpaired two-tailedlevels of typical postsynapticStudent’s t-test; genesn = 3were cell sets). significantly reduced (**** p < 0.0001, *** p < 0.001, ** p < 0.01; * p < 0.05; unpaired two-tailed Student’s t-test; n = 3 cell sets). 3.5. The Turnover of Neurotransmitter Receptor Clusters Is Impressively Increased in CK2β-Deficient Muscles

3.5. TheWe Turnover asked whether of Neurotransmitter the absence of Recept CK2β orinfluenced Clusters AChRis Impressively stability, asIncreased was suggested in CK2β before-Deficient by the Muscles disappearance of agrin-induced AChR clusters after the withdrawal of agrin but in the presence of CK2We inhibitors asked whether in cell culture the absence [8]. Here, of weCK2 decidedβ influenced to monitor AChR the stability, AChR turnover as was in suggested CK2β-deficient before by the disappearancemuscles directly. of agrin-induced Therefore, tibialis AChR anterior clusters (TA) after muscles the withdrawal of control and of CK2agrinβ-deficient but in the mice presence were of CK2 inhibitorsinjected with in cell BTX-AF647. culture [8]. Ten Here, days we later, de thecided muscles to monitor were injected the AChR with turnover BTX-AF555. in Then,CK2β BTX-AF647-deficient muscles directly. Therefore, tibialis anterior (TA) muscles of control and CK2β-deficient mice were injected with BTX-AF647. Ten days later, the muscles were injected with BTX-AF555. Then, BTX-AF647 (old AChR) and BTX-AF555 (new AChR) fluorescence signals were monitored by in vivo confocal microscopy. Previously, it was reported that, given the extreme stability of the AChR-BTX complex and the small quantities of BTX (25 pmol) injected at each occasion, the largest proportion of BTX should have been available in the circulatory system only for a limited amount of time. Therefore, AChRs marked by BTX should have been surface-exposed at the time point of injection, i.e., ten days or about one hour before

Cells 2019, 8, 940 12 of 16

(old AChR) and BTX-AF555 (new AChR) fluorescence signals were monitored by in vivo confocal microscopy. Previously, it was reported that, given the extreme stability of the AChR-BTX complex and the small quantities of BTX (25 pmol) injected at each occasion, the largest proportion of BTX should have been available in the circulatory system only for a limited amount of time. Therefore, AChRs marked by BTX should have been surface-exposed at the time point of injection, i.e., ten days or about one hourCells 2019 before, 8, x inspection for BTX-AF647 or BTX-AF555, respectively [27]. In control muscles,13 of the17 two BTX fluorescence signals showed a very significant overlap and clearly depicted the typical arborized inspection for BTX-AF647 or BTX-AF555, respectively [27]. In control muscles, the two BTX fluorescence structuresignals of showed adult NMJsa very significant (Figure7A). overlap In CK2 and βclearl-deficienty depicted muscles, the typical the arborized overall architecture structure of adult of NMJs was significantlyNMJs (Figure 7A). altered In CK2 andβ-deficient appeared muscles, fragmented the overall (Figure architec7B).ture Furthermore, of NMJs was althoughsignificantly the altered intensity and theand extensionappeared fragmented of the “new (Figure receptors” 7B). Furthermore, were still comparablealthough the intensity to the control and the muscles extension (Figure of the 7A), “old receptors”“new receptors” were were completely still comparable gone in to CK2 theβ -deficientcontrol muscles muscles (Figure (Figure 7A),7 B).“old These receptors” changes were were reflectedcompletely by the gone quantities in CK2β-deficient of “old” muscles and “new” (Figure receptors 7B). These in changes the muscles were reflected (Figure by7 theC,D). quantities While the amountof “old” of BTX-AF647 and “new” wasreceptors lowered in the in CK2musclesβ-deficient (Figure 7C,D). NMJs While (Figure the7 C),amount the amount of BTX-AF647 of BTX-AF555 was was significantlylowered in CK2 increasedβ-deficient in NMJs CK2β (Figure-deficient 7C), NMJs the amount (Figure of7 BTX-AF555D). In summary, was significantly analyzed increased CK2 β-deficient in CK2β-deficient NMJs (Figure 7D). In summary, analyzed CK2β-deficient NMJs displayed both a higher NMJs displayed both a higher fragmentation and an increased AChR turnover rate. fragmentation and an increased AChR turnover rate.

FigureFigure 7. NMJs 7. NMJs became became fragmented fragmented and and showedshowed a high turnover turnover of ofAChRs AChRs in CK2 in CK2β-deficientβ-deficient muscles. muscles. (A,B)(AIn,B vivo) In vivomicroscopy microscopy and and analysis analysis ofof AChR turn turnoverover and and NMJ NMJ fragmentation fragmentation in soleus in soleusmuscle muscleof control (A) and conditional CK2β-deficient (B) mice. TA muscles of 6–9 months old control and of control (A) and conditional CK2β-deficient (B) mice. TA muscles of 6–9 months old control and conditional CK2β-deficient mice were studied. Simultaneously, muscles were locally injected with BTX- conditional CK2β-deficient mice were studied. Simultaneously, muscles were locally injected with AF647. Ten days later, muscles were locally injected with BTX-AF555 and then imaged using in vivo BTX-AF647. Ten days later, muscles were locally injected with BTX-AF555 and then imaged using confocal microscopy. Maximum z-projections of 45 (CK2β-deficient, B) and 55 (control, A) optical slices in vivowereconfocal taken at microscopy. 1.5 µm interslice Maximum distance encompassing z-projections the of entire 45 (CK2 exteβnsions-deficient, of shownB) andNMJs 55 in (control,depth. A) opticalOverlay slices image, were takenBTX-AF647 at 1.5 (green),µm interslice BTX-AF555 distance (red), colocalization encompassing (yellow). the entire Scale extensionsbar, 50 µm. ( ofC,D shown) NMJsThese in depth. panels Overlay show the image, comparison BTX-AF647 of mean (green), intensity BTX-AF555 of either BTX-AF647 (red), colocalization or BTX-AF555 (yellow). labeled NMJs Scale bar, 50 µm.in control (C,D) Theseand wild-type panelsshow and CK2 theβ comparison-deficient muscles. of mean (**** intensity p < 0.0001; of * either p < 0.05; BTX-AF647 unpaired two-tailed or BTX-AF555 labeledStudent’s NMJs int-test; control >15 NMJs and wild-typewere analyzed and of CK2 bothβ-deficient control and muscles. CK2β-deficient (**** p muscles.< 0.0001; * p < 0.05; unpaired two-tailed Student’s t-test; >15 NMJs were analyzed of both control and CK2β-deficient muscles. 4. Discussion The protein kinase CK2 was shown to play a substantial role in adult muscle fibers and at NMJs by interacting and/or phosphorylating members of the protein import machinery of mitochondria or members of the postsynaptic apparatus [8,12,16,17]. Since we used a conditional CK2β-deficient mouse model and ensured proper deletion of CK2β by HSA-Cre reporter mice, it is likely that the changes we observed in the mutant mice took place in an age-dependent manner at more than 2–3

Cells 2019, 8, 940 13 of 16

4. Discussion The protein kinase CK2 was shown to play a substantial role in adult muscle fibers and at NMJs by interacting and/or phosphorylating members of the protein import machinery of mitochondria or members of the postsynaptic apparatus [8,12,16,17]. Since we used a conditional CK2β-deficient mouse model and ensured proper deletion of CK2β by HSA-Cre reporter mice, it is likely that the changes we observed in the mutant mice took place in an age-dependent manner at more than 2–3 months. In fact, our studies targeted the role of CK2 in adult muscle fibers and did not analyze the influence of CK2 in early myogenesis or injury-induced muscle regeneration. Here, we wanted to explore the structural and the functional links between CK2 and NMJs in more detail. The differences in neuromuscular transmission between CK2β-deficient muscles and controls appeared to be rather small, correlated with the same ability of mutant mice regarding voluntary walking controls in an age-independent manner [16]. Yet, we detected a significant loss of grip-strength in conditional CK2β-deficient mice in comparison with controls (Figure2A,B), as well as a higher degree of fatigue (Figure2B,D). These behavioral changes in mutant mice were accompanied by structural and functional changes. First, we detected a higher fragmentation grade of NMJs in mutants in comparison with controls (Figures4 and5). Second, we recorded lower amplitudes of miniature endplate potentials and currents (Figure1B,C), and higher fatigability, as evidenced by lower decrements of endplate potential amplitudes after run-down experiments (Figure3B). Neuromuscular transmission impairments might have been masked by the safety factor [31]. Remarkably, we used our established ex vivo electrophysiological recording setup for measurements in the presence of increasing tubocurarine concentration to detect a prominent and significant decrease of the decrements of the compound muscle action potential in mutant diaphragms in comparison with controls (Figure3D) [ 36]. Third, further information was also provided on the distribution and the size of differentially labeled pools of AChRs (Figures4 and7). BTX stainings of newly arriving or old receptors showed their highest densities at the rim or the center of NMJs, respectively [37]. For NMJs of CK2β-deficient muscles, fluorescence signals of old receptors disappeared, arguing for a very high turnover of AChR clusters in mutant mice. We wondered what the reasons for the rise of the AChRs turnover were. Presumably, the proven phosphorylation of the postsynaptic proteins MuSK, Dok-7, and 14-3-3γ was functionally significant at NMJs [8,12]. However, what about the expression profiles of postsynaptic genes? They might also have been compromised, either due to direct influences on their expression or because of regulatory compensation reactions. The significant increase of myonuclei diameter in muscle fibers of adult CK2β-deficient mice already pointed to an eventual increase of transcriptional activity (Figure6A). Indeed, we detected a strong increase of transcriptional activities of AChR subunits α, β, δ, and ε (Figure6B) in addition to AChR γ, although this increase might have been more related to regeneration events rather than a direct consequence of CK2β ablation (Figure6C). This increase in the transcriptional rate of AChRs was in agreement with the amount of AChR protein (Figure7D). The quantity of the new receptor (BTX-AF555) was significantly increased in NMJs of CK2β-deficient muscles (Figure7D). Interestingly, transcript levels of MuSK were also increased, while Rapsyn transcription was not affected (Figure6D,E). These findings might explain why MuSK was detected being phosphorylated by CK2 but not Rapsyn [8,12]. On the other hand, the transcript levels of Rapsyn also appeared to be a little lower in CK2β-deficient muscles in comparison with controls, although these decreases were not statistically significant (Figure6E). Unexpectedly, transcript levels of postsynaptic genes were lower in cultured myotubes, which were incubated with two different CK2 inhibitors, apigenin and DMAT (Figure6F). This discrepancy between the expression levels of postsynaptic genes in CK2 β-deficient muscles and cultured muscle cells in the presence of CK2 inhibitors might be explainable, keeping in mind the different turnover times of clustered AChRs of 4–8 h in vitro in cultured myotubes in comparison to two weeks in vivo. In this study, we did not want to analyze aging or functional denervation defects in the absence of CK2β in skeletal muscles. However, our data thus far showed no apparent change in the presynaptic Cells 2019, 8, 940 14 of 16 compartment in CK2β-deficient muscles, which speaks against denervation defects. Nevertheless, functional denervation would be conceivable. Overall, our data indicate that CK2 activity on NMJs is required for physiological AChR expression and AChR cluster turnover.

Author Contributions: Conceptualization, S.H.; methodology, N.E., M.R., B.K.; validation, S.H., N.E., M.S., R.R.; formal analysis, S.H., N.E., M.R.; investigation, N.E., B.K., M.R.; resources, S.H., M.S., R.R.; data curation, S.H., N.E., M.R.; writing—original draft preparation, S.H.; writing—review and editing, S.H., N.E., M.R., M.S., R.R.; supervision, S.H.; project administration, S.H.; funding acquisition, S.H., M.S., R.R. Funding: This work was supported by the German Research Council DFG to SH (HA 3309/1-3 & HA3309/3-1) and RR (RU923/8-2). Further support was obtained from the, Johannes und Frieda Marohn-Stiftung, and the Interdisciplinary Centre for Clinical Research at the University Hospital of the Friedrich-Alexander University of Erlangen-Nürnberg (E2 & E17), all to SH. Conflicts of Interest: The authors declare no conflict of interest.

References

1. Li, L.; Xiong, W.C.; Mei, L. Neuromuscular Junction Formation, Aging, and Disorders. Annu. Rev. Physiol 2018, 80, 159–188. [CrossRef][PubMed] 2. Weatherbee, S.D.; Anderson, K.V.; Niswander, L.A. LDL-receptor-related protein 4 is crucial for formation of the neuromuscular junction. Development 2006, 133, 4993–5000. [CrossRef][PubMed] 3. Zhang, B.; Luo, S.; Wang, Q.; Suzuki, T.; Xiong, W.C.; Mei, L. LRP4 serves as a coreceptor of agrin. Neuron 2008, 60, 285–297. [CrossRef][PubMed] 4. Kim, N.; Stiegler, A.L.; Cameron, T.O.; Hallock, P.T.; Gomez, A.M.; Huang, J.H.; Hubbard, S.R.; Dustin, M.L.; Burden, S.J. Lrp4 is a receptor for Agrin and forms a complex with MuSK. Cell 2008, 135, 334–342. [CrossRef] [PubMed] 5. DeChiara, T.M.; Bowen, D.C.; Valenzuela, D.M.; Simmons, M.V.; Poueymirou, W.T.; Thomas, S.; Kinetz, E.; Compton, D.L.; Rojas, E.; Park, J.S.; et al. The receptor tyrosine kinase MuSK is required for neuromuscular junction formation in vivo. Cell 1996, 85, 501–512. [CrossRef] 6. Glass, D.J.; DeChiara, T.M.; Stitt, T.N.; DiStefano, P.S.; Valenzuela, D.M.; Yancopoulos, G.D. The receptor tyrosine kinase MuSK is required for neuromuscular junction formation and is a functional receptor for agrin. Cold Spring Harb. Symp. Quant. Biol. 1996, 61, 435–444. [PubMed] 7. Simeone, L.; Straubinger, M.; Khan, M.A.; Nalleweg, N.; Cheusova, T.; Hashemolhosseini, S. Identification of Erbin interlinking MuSK and ErbB2 and its impact on acetylcholine receptor aggregation at the neuromuscular junction. J. Neurosci. 2010, 30, 6620–6634. [CrossRef][PubMed] 8. Cheusova, T.; Khan, M.A.; Schubert, S.W.; Gavin, A.C.; Buchou, T.; Jacob, G.; Sticht, H.; Allende, J.; Boldyreff, B.; Brenner, H.R.; et al. Casein kinase 2-dependent serine phosphorylation of MuSK regulates acetylcholine receptor aggregation at the neuromuscular junction. Genes Dev. 2006, 20, 1800–1816. [CrossRef] 9. Okada, K.; Inoue, A.; Okada, M.; Murata, Y.; Kakuta, S.; Jigami, T.; Kubo, S.; Shiraishi, H.; Eguchi, K.; Motomura, M.; et al. The muscle protein Dok-7 is essential for neuromuscular synaptogenesis. Science 2006, 312, 1802–1805. [CrossRef] 10. Luo, Z.G.; Wang, Q.; Zhou, J.Z.; Wang, J.; Luo, Z.; Liu, M.; He, X.; Wynshaw-Boris, A.; Xiong, W.C.; Lu, B.; et al. Regulation of AChR clustering by Dishevelled interacting with MuSK and PAK1. Neuron 2002, 35, 489–505. [CrossRef] 11. Luo, Z.G.; Je, H.S.; Wang, Q.; Yang, F.; Dobbins, G.C.; Yang, Z.H.; Xiong, W.C.; Lu, B.; Mei, L. Implication of geranylgeranyltransferase I in synapse formation. Neuron 2003, 40, 703–717. [CrossRef] 12. Herrmann, D.; Straubinger, M.; Hashemolhosseini, S. Protein kinase CK2 interacts at the neuromuscular synapse with Rapsyn, Rac1, 14-3-3gamma, and Dok-7 proteins and phosphorylates the latter two. J. Biol. Chem. 2015, 290, 22370–22384. [CrossRef] 13. Litchfield, D.W. Protein kinase CK2: Structure, regulation and role in cellular decisions of life and death. Biochem. J. 2003, 369, 1–15. [CrossRef][PubMed] 14. Buchou, T.; Vernet, M.; Blond, O.; Jensen, H.H.; Pointu, H.; Olsen, B.B.; Cochet, C.; Issinger, O.G.; Boldyreff, B. Disruption of the regulatory beta subunit of protein kinase CK2 in mice leads to a cell-autonomous defect and early embryonic lethality. Mol. Cell. Biol. 2003, 23, 908–915. [CrossRef][PubMed] Cells 2019, 8, 940 15 of 16

15. Seldin, D.C.; Lou, D.Y.; Toselli, P.; Landesman-Bollag, E.; Dominguez, I. Gene targeting of CK2 catalytic subunits. Mol. Cell. Biochem. 2008, 316, 141–147. [CrossRef][PubMed] 16. Kravic, B.; Harbauer, A.B.; Romanello, V.; Simeone, L.; Vogtle, F.N.; Kaiser, T.; Straubinger, M.; Huraskin, D.; Bottcher, M.; Cerqua, C.; et al. In mammalian skeletal muscle, phosphorylation of TOMM22 by protein kinase CSNK2/CK2 controls mitophagy. Autophagy 2018, 14, 311–335. [CrossRef][PubMed] 17. Eiber, N.; Simeone, L.; Hashemolhosseini, S. Ablation of Protein Kinase CK2beta in Skeletal Muscle Fibers Interferes with Their Oxidative Capacity. Pharmaceuticals 2017, 10, 13. [CrossRef][PubMed] 18. Heuss, D.; Klascinski, J.; Schubert, S.W.; Moriabadi, T.; Lochmuller, H.; Hashemolhosseini, S. Examination of transcript amounts and activity of protein kinase CK2 in muscle lysates of different types of human muscle pathologies. Mol. Cell. Biochem. 2008, 316, 135–140. [CrossRef][PubMed] 19. Toyka, K.V.; Brachman, D.B.; Pestronk, A.; Kao, I. Myasthenia gravis: Passive transfer from man to mouse. Science 1975, 190, 397–399. [CrossRef] 20. Ruvinsky, I.; Katz, M.; Dreazen, A.; Gielchinsky, Y.; Saada, A.; Freedman, N.; Mishani, E.; Zimmerman, G.; Kasir, J.; Meyuhas, O. Mice deficient in ribosomal protein S6 phosphorylation suffer from muscle weakness that reflects a growth defect and energy deficit. PLoS ONE 2009, 4, e5618. [CrossRef] 21. Liley, A.W. An investigation of spontaneous activity at the neuromuscular junction of the rat. J. Physiol. 1956, 132, 650–666. [CrossRef] 22. Sandrock, A.W., Jr.; Dryer, S.E.; Rosen, K.M.; Gozani, S.N.; Kramer, R.; Theill, L.E.; Fischbach, G.D. Maintenance of acetylcholine receptor number by neuregulins at the neuromuscular junction in vivo. Science 1997, 276, 599–603. [CrossRef] 23. Plomp, J.J.; van Kempen, G.T.; Molenaar, P.C. Adaptation of quantal content to decreased postsynaptic sensitivity at single endplates in alpha-bungarotoxin-treated rats. J. Physiol. 1992, 458, 487–499. [CrossRef] [PubMed] 24. Rogozhin, A.A.; Pang, K.K.; Bukharaeva, E.; Young, C.; Slater, C.R. Recovery of mouse neuromuscular junctions from single and repeated injections of botulinum neurotoxin A. J. Physiol. 2008, 586, 3163–3182. [CrossRef][PubMed] 25. Kroger, S. Differential distribution of agrin isoforms in the developing and adult avian retina. Mol. Cell. Neurosci. 1997, 10, 149–161. [CrossRef] 26. Gesemann, M.; Denzer, A.J.; Ruegg, M.A. Acetylcholine receptor-aggregating activity of agrin isoforms and mapping of the active site. J. Cell. Biol. 1995, 128, 625–636. [CrossRef][PubMed] 27. Roder, I.V.; Petersen, Y.; Choi, K.R.; Witzemann, V.; Hammer, J.A.; Rudolf, R. Role of Myosin Va in the plasticity of the vertebrate neuromuscular junction in vivo, 3rd ed. PLoS ONE 2008, 3, e3871. [CrossRef] [PubMed] 28. Yampolsky, P.; Pacifici, P.G.; Lomb, L.; Giese, G.; Rudolf, R.; Roder, I.V.; Witzemann, V. Time lapse in vivo visualization of developmental stabilization of synaptic receptors at neuromuscular junctions. J. Biol. Chem. 2010, 285, 34589–34596. [CrossRef] 29. Kravic, B.; Huraskin, D.; Frick, A.D.; Jung, J.; Redai, V.; Palmisano, R.; Marchetto, S.; Borg, J.P.; Mei, L.; Hashemolhosseini, S. LAP proteins are localized at the post-synaptic membrane of neuromuscular junctions and appear to modulate synaptic morphology and transmission. J. Neurochem. 2016, 139, 381–395. [CrossRef] 30. Banker, B.Q.; Kelly, S.S.; Robbins, N. Neuromuscular transmission and correlative morphology in young and old mice. J. Physiol. 1983, 339, 355–377. [CrossRef] 31. Wood, S.J.; Slater, C.R. The contribution of postsynaptic folds to the safety factor for neuromuscular transmission in rat fast- and slow-twitch muscles. J. Physiol. 1997, 500, 165–176. [CrossRef][PubMed] 32. Andonian, M.H.; Fahim, M.A. Effects of endurance exercise on the morphology of mouse neuromuscular junctions during ageing. J. Neurocytol. 1987, 16, 589–599. [CrossRef][PubMed] 33. Rudolf, R.; Khan, M.M.; Labeit, S.; Deschenes, M.R. Degeneration of Neuromuscular Junction in Age and Dystrophy. Front. Aging Neurosci. 2014, 6, 99. [CrossRef][PubMed] 34. Webster, M.; Witkin, K.L.; Cohen-Fix, O. Sizing up the nucleus: Nuclear shape, size and nuclear-envelope assembly. J. Cell. Sci. 2009, 122, 1477–1486. [CrossRef][PubMed] 35. Witzemann, V.; Barg, B.; Criado, M.; Stein, E.; Sakmann, B. Developmental regulation of five subunit specific mRNAs encoding acetylcholine receptor subtypes in rat muscle. FEBS Lett. 1989, 242, 419–424. [CrossRef] Cells 2019, 8, 940 16 of 16

36. Cescon, M.; Gregorio, I.; Eiber, N.; Borgia, D.; Fusto, A.; Sabatelli, P.; Scorzeto, M.; Megighian, A.; Pegoraro, E.; Hashemolhosseini, S.; et al. Collagen VI is required for the structural and functional integrity of the neuromuscular junction. Acta Neuropathol. 2018, 136, 483–499. [CrossRef][PubMed] 37. Yampolsky, P.; Gensler, S.; McArdle, J.; Witzemann, V. AChR channel conversion and AChR-adjusted neuronal survival during embryonic development. Mol. Cell. Neurosci. 2008, 37, 634–645. [CrossRef]

© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).