Abstract
Myocardial infarction (MI) is a leading cause of death globally and loss of cardiomyocytes plays a critical role in the pathogenesis of heart failure. Implicated in this process is reduced gap junction intercellular communication due to remodeling of Connexin43 (Cx43). We previously identified that intraperitoneal injection of the Pyk2 inhibitor PF4618433, reduced infarct size, maintained Cx43 at the intercalated disc in left ventricle hypertrophic myocytes, and improved cardiac function in a MI animal model of heart failure. With the emergence of injectable hydrogels as a therapeutic towards regeneration of cardiac tissue after MI, here we provide proof of concept that release of kinase inhibitors from hydrogels could have beneficial effects on cardiomyocytes. We developed an injectable hydrogel consisting of thiolated hyaluronic acid and P123-maleimide micelles, that can incorporate PF4618433 as well as the Src inhibitor Saracatinib and achieved sustained release. Using neonatal rat ventricular myocytes in the presence of a phorbol ester, endothelin-1, or phenylephrine to stimulate cardiac hypertrophy, release of PF4618433 from the hydrogel had the same ability to decrease Cx43 tyrosine phosphorylation and maintain Cx43 localization at the plasma membrane as when directly added to the growth media. Additional beneficial effects included decreases in apoptosis, the hypertrophic marker Atrial natriuretic peptide (ANP), and serine kinases upregulated in hypertrophy. Finally, presence of both PF4618433 and Saracatinib further decreased the level of ANP and apoptosis than each inhibitor alone, suggesting a combinatorial approach may be most beneficial. These findings provide the groundwork to test if tyrosine kinase inhibitor release from hydrogels will have a beneficial effect in an animal model of MI induced heart failure.
Keywords: injectable hydrogel, tyrosine kinase inhibitor, Cx43, gap junctions, Src, Pyk2
Graphical Abstract
1. Introduction
Gap junction channels composed of Connexin43 (Cx43) mediate electrical coupling and impulse propagation in the normal working myocardium. Following a myocardial infarction (MI), the cardiac muscle sarcoplasm from the surviving ventricular myocytes (epicardial border zone), experience many abnormalities, including reduced gap junction intercellular communication due to changes in Cx43 expression, phosphorylation state, and increased relocalization from the intercalated disc to the lateral membrane1–7. Reduced gap junctional coupling causes instability of action potential duration and refractory period, effects seen in the epicardial border zone8.
Left ventricle hypertrophy following a MI is an adaptive response to increased biomechanical stress9. Initially, heart mass increases to normalize wall stress and retain normal cardiovascular function, accompanied by an increase in Cx43 expression and gap junction intercellular communication10. However, the increased cardiac mass and sustained overload eventually lead to contractile dysfunction and heart failure11. As left ventricle hypertrophy becomes severe, propagation velocity decreases, which correlates with reduced Cx43 expression, altered phosphorylation which affects channel regulation and protein partners interactions, and translocation from the intercalated disc to the lateral membrane12–14. While Cx43 lateralization is an almost ubiquitous response to cardiac pathology8, 15, 16, the molecular mechanisms of this remodeling are ill-defined. Based upon data from our lab and others, a model was put forth describing a mechanism for laterization involving Src and Pyk2 phosphorylation of Cx43 causing dissociation of ZO-1 (binds F-actin), β-tubulin, and Drebrin (binds F-actin)7, 17–19. Initially, ZO-1 unhooks from Cx43 causing the plaque to increase in size20, 21. Then as it is no longer anchored by the cytoskeletal network, Cx43 moves away from the region of high concentration at the intercalated disc to the lateral membrane7, 22. Lateralization is a key factor that reduces conduction leading to reentrant arrhythmias8, 23.
Unlike other attempts to decrease Cx43 remodeling13, 24, 25, inhibition of Pyk2 activity in an animal model of heart failure using the small molecule inhibitor PF4618433 maintained Cx43 at the intercalated disc in left ventricle hypertrophic myocytes and improved cardiac function26. Additionally, although the acute time frame was not mechanistically investigated, PF4618433 also reduced the infarct size26. PF4618433 is a diaryl urea small molecule inhibitor that binds Pyk2 in an allosteric binding pocket distinct from the ATP pocket to help minimizing off-target activity27. In comparison to classical Pyk2 inhibitors, PF4618433 had superior overall selectivity (>35 kinases tested) and improved potency to other allosteric binding pocket Pyk2 inhibitors with an IC50 of 637 nM27. Pyk2 is activated following MI in human left ventricle implicating a novel potential target for therapy in patients with heart failure26. Of note, the Src inhibitor Saracatinib was also included in this study because Src directly activates Pyk214. In an animal model, Saracatinib decreased Src activity to the sham level, raised Cx43 expression at the scar border and distal ventricle, and had a higher level of the Cx43 P2 isoform (correlates with promoting gap junction intercellular communication)13. While no decrease in lateralization was observed, this still led to ~50% improvement in conduction velocity and lowered arrhythmia inducibility13.
Current available treatments of MI include pharmaceutical therapy, medical device implants, and organ transplants, however they all have their inherent limitations28–30. Injectable hydrogels have emerged as a promising translational therapeutic towards regeneration of cardiac tissue after MI. These hydrogels have the capacity to encapsulate cells and/or locally release therapeutics after injection through a syringe or catheter31, 32. Several formulations for injection into cardiac tissue have been demonstrated as safe in animals and are in clinical trials33, 34. Recent hydrogels engineered for cardiac tissue to reverse the adverse myocardial microenvironment include matrix metalloproteinase-responsive hydrogels, reactive oxygen species-scavenging hydrogels, and immunomodulatory hydrogels35. Hydrogels have been designed to improve electrical signal conduction and vascularization within the infarct area to promote the repair of cardiac function. Finally, 3D-printed hydrogels, which can achieve personal customized cardiac tissue via printing of intact cardiac structures, were developed to address the current shortage of heart donors36. Here, we developed a micelle crosslinked hydrogel and tested the release of Pyk2 and Src inhibitors that have shown to be beneficial after a MI (in acute ischemia and in late stage left ventricle hypertrophy), however the mode of delivery was intraperitoneal injection. For this in vitro proof of concept study, we tested three different reagents phorbol 12-myristate 13-acetate (PMA), endothelin-1 (ET-1), and phenylephrine. ET-1 and phenylephrine are activated during ischemic injury and heart failure37–40. All three induce cardiac cell hypertrophy and re-expression of cardiac-specific fetal genes14, 41–46. We anticipate that our hydrogel system will be an improved delivery vehicle because the amount of inhibitor needed will be less, targets only the cardiac tissue, and allows for slow release over a long period of time.
2. Materials and methods
2.1. Synthesis of thiolated hyaluronic acid (HA-SH) and P123-maleimide (PM)
The HA-SH polymer conjugate was prepared by conjugating cysteamine (Sigma) to HA (290 kDa, Bloomage Biotech) using 1-ethyl-3-(3-dimethyl aminopropyl) carbodiimide (EDC)/N-hydroxysuccinimide (NHS) as the coupling agent (Fig. 1)47. In brief, 200 mg of HA (0.5 mmol) was dissolved in 20 mL of de-ionized (DI) water and was activated after adding 384 mg of EDC (2 mmol, Oakwood Chemical) and 115 mg of NHS (1 mmol, Oakwood Chemical) under constant stirring. Next, 230 mg of cysteamine (2 mmol) was poured into the above mixture. Then the pH of the solution was adjusted to 5.0 using a 1N HCl (Fisher Chemical) solution. The reaction mixture was stirred at room temperature (RT) for 24 hr, transferred to a dialysis bag with a 6–8 kDa molecular weight cut-off (MWCO, Spectrum) and dialyzed against acidic DI water (with the addition of 1 mM tris(2-carboxyethyl)phosphine or TCEP at pH 3) for 3 days at RT, with the water changed every 12 hr. The purified conjugate solution was then neutralized with sodium hydroxide solution (1N NaOH, Fisher) and freeze-dried in a benchtop lyophilizer (model FreeZone, Labconco) for 2 days to acquire the purified HA-SH polymer conjugate. The conjugates were stored at −80°C.
Figure 1.
Schematic illustration of the micelle crosslinked injectable hydrogels for dual TKI delivery to cardiomyocytes.
The PM conjugate was synthesized in two steps. In the first step, 580 mg of Pluronic P123 (0.1 mmol, Sigma) and 122 mg of 4-dimethylaminopyridine (DMAP, 1 mmol, Sigma) were dissolved in 2 mL of 1,4-dioxane (Sigma) with 139 μL of triethylamine (TEA). After stirring at RT for 15 min, 200 mg of succinic anhydride (2 mmol, Sigma) in 2 ml of 1,4-dioxane was added to the Pluronic P123 solution48. The mixture was stirred at RT for 24 hr and then the organic solvent was removed in vacuum. The residual sample was precipitated in ice-cold ether (EMD Millipore) twice. The precipitate was dissolved in water and then freeze-dried to obtain the white powder of dicarboxylated Pluronic P123 (P123-succinic acid). In the second step, after dissolving 100 mg of P123-succinic acid (17 μmol) in 5 mL of ice-cold dichloromethane (DCM), 23 mg of 1-Hydroxybenzotriazole (HOBt, 170 μmol, Oakwood) in 100 μL of dimethyl sulfoxide (DMSO, Fisher) was added dropwise and the mixture was stirred for 10 min. Next, 15 mg of N-(2-aminoethyl)maleimide hydrochloride (85 μmole TCI) in 100 μL of DMSO and 10 μL of TEA were added respectively. After stirring for another 15 min, 32.6 mg of EDC (170 μmol) in 300 μL of DMSO was added to the solution and the final mixture was stirred at RT for 24 hr. DCM was removed by vacuum and the residual was solubilized in water. The reaction mixture solution was dialyzed against water for 3 days and the purified PM was lyophilized and kept at −20°C before use.
2.2. Proton nuclear magnetic resonance (1H NMR) analysis of polymers
1H NMR characterization was performed on a 500 MHz Bruker NMR system and analyzed with Topspin 4.0 software. The polymer conjugate was dissolved in D2O (Acros Organic) at 5 mg/mL for NMR acquisition, with the chemical shifts referring to the solvent peak of D2O at 4.78 ppm at 25°C. The substitution degree of HA-SH was determined from the ratio of the integral of ethyl protons from the conjugated cysteamine (between 2.2~2.4 ppm, 4H, -C2H4) to the integral of the HA methyl proton peak (at 1.9 ppm, 3H, -CH3). The grafting ratio of PM was calculated from the ratio of the integral of the alkene protons from the attached maleimide group (between 6.7~6.9 ppm, 2*C2H2) to the integral of P123 methyl proton peak (~1.1 ppm, 70*CH3).
2.3. Preparation and rheological characterization of HA-PM hydrogels
An equal volume of 5% (w/v) PM dissolved in water was quickly mixed with either 1% or 2% (w/v) HA-SH in water to prepare the HA-PM hydrogels and the two hydrogels were referred to as the 5:1 and 5:2 HA-PM hydrogel, respectively49. To investigate the gelation behavior, rheological studies were conducted. The rheological properties of the HA-PM hydrogels were characterized at 37°C using a Discovery HR-2 rheometer (TA Instruments) with several different protocols50. In each study, around 0.2 mL of total hydrogel precursor solution was mixed in an Eppendorf tube and thus quickly transferred into the geometry gap between the 20 mm parallel plate and the base plate. The time sweep method was carried out with a fixed angular frequency of 2π rad/s (=1 Hz) and 10% strain to measure the storage modulus (G’) and loss moduli (G”) over time. Frequency sweeps from 0.1 to 10 Hz were performed at 10% constant strain to validate the hydrogel viscoelasticity. Flow sweep study was conducted on the hydrogels with a linearly ramped shear rate from 1 to 100 s–1, and the viscosity recorded at different rates.
2.4. Swelling and degradation study
Each piece of hydrogel (50 μL) was incubated in 1 mL of PBS (Fisher, pH 7.4) in a 1.5 mL Eppendorf tube at 37°C. At each time point, hydrogel was taken out from the PBS, weighed, and then transferred into 1 mL of fresh PBS50. Weight of each hydrogel sample (Ws) was determined after removing excess PBS from the hydrogel surface. The initial weight was set as W0. Hydrogel swelling was monitored over 24 hr and calculated with the equation: Ws/W0×100%. The degradation ratio was calculated using the equation: (W0-Ws)/W0×100%. Each group had three replicates.
2.5. Primary cardiomyocyte culture and MTT study
The animal procedure was approved by the institutional animal care and use committee at the University of Nebraska Medical Center. Primary cardiomyocytes (neonatal rat ventricular myocytes, NRVMs) were isolated from day 1–3 neonatal rat hearts using the Pierce Primary Cardiomyocyte Isolation Kit (>90% pure) per manufacturer protocol (catalog number: 88281)14. NRVMs were plated at a seeding density of 2.5 × 105 cells/cm2 in complete DMEM supplemented with 10% FBS and 1% pen-strep at 37°C in a 5% CO2 incubator for 24 hr. The medium was then replaced with complete DMEM containing Cardiomyocyte Growth Supplement for 3 days. A piece of hydrogel (50 μL) was then added to each well of cells. The control group contained no hydrogel. Each group had 3 replicates. After 2 days incubation, the cytotoxicity was determined using MTT (Sigma) assay with the absorbance measured at 540 nm in a microplate reader (BioTek)14. The absorbance value from each group was normalized to the average absorbance value in the control group.
2.6. Preparation of P123 micelles loaded with dual tyrosine kinase inhibitors (TKIs)
Both Src and Pyk inhibitors are hydrophobic and show limited water solubility. PM micelles were applied to solubilize the drugs. The micelles were prepared by thin film hydration method49. Generally, both 5% (w/v) PM conjugate was first dissolved in methanol. TKI stock solution was also prepared in methanol. The PM methanol was then added to aliquot amount of TKIs in an Eppendorf tube. After complete mixing, the methanol was evaporated in vacuum and a thin film was formed on the tube surface. Next an equal volume of DI water was added to hydrate the film for at least 15 min at RT. The mixture was sonicated for another 15 min and the non-encapsulated TKIs were removed by centrifugation at 10,000 g for 5 min. The size and polydispersity (PdI) of the micelles encapsulated with each drug was determined by dynamic light scattering measurement (Malvern). The non-encapsulated TKIs were dissolved in 100 μL of methanol and injected into high performance liquid chromatography (HPLC) to determine the drug concentration (method described below). The encapsulation (EE) was calculated by the equation: EE = (Weight of feeding drug - Weight of non-encapsulated drug) / Weight of feeding drug × 100%.
2.7. FTIR characterization
The polymer precursor, blank hydrogel, free drug, drug loaded micelle and drug loaded hydrogels were analyzed using a Nicolet Is50 FT-IR spectrometer (Thermo Scientific) in the range 500–4000 cm−1. The data was recorded with the OMNIC software. The drug/polymer/lyophilized hydrogel powder was mixed with KBr and subjected to scanning.
2.8. Scanning electron microscopy analysis
The HA-SH were crosslinked to the drug loaded micelles to prepare TKIs loaded hydrogels. The micro-structure of the freeze-dried HA-PM hydrogel was studied by scanning electron microscopy (SEM, FEI Quanta 200). The freshly prepared drug loaded HA-PM hydrogels were completely lyophilized and then sprayed on a thin film of gold. The sample was then transferred to the SEM instrument for examination.
2.9. HPLC analysis
The release of both drugs from the TKI loaded hydrogels was determined by the dialysis method in PBS solution. The released amount of individual drug at each time point was simultaneously quantified by the Agilent 1260 HPLC at 250 nm equipped with a diode array detector by injecting 30 μL of the releasate into the HPLC instrument. A Poroshell 120 EC-C-18 column (3.0 × 150 mm, 2.7 μm particle size) was used for drug separation and quantification at 30°C and 0.5 mL/min flow rate14. The mobile phase consisted of an aqueous phase (A) containing 0.1% (v/v) formic acid and an organic phase (B) containing acetonitrile. A gradient method was used, in which the mobile phase started as 80% A and 20% B and linearly changed to 20% A and 80% B in 8 min. The mobile phase ratio was held consistent between 8 to 10 min and was switched to 5% A and 95% B by the12th min. It steadily increased to 80% A and 20% B by the 15th min. A standard curve was generated with known concentrations of TKIs (1–10 μg/mL) prepared in water/ACN (50:50 v/v). The percentage of drug release was calculated by dividing the accumulative amount of released drugs to the total encapsulated drugs at the beginning.
2.10. Pyk2 and Src inhibitor treatment
After purification and seeding (see MTT section above for more details), NRVMs were pre-treated with PF4618433, Saracatinib, or both (either dissolved in medium or incorporated within hydrogels) for 3 hr and then either PMA, ET-1, or Phenylephine was added for 1 hr or 24 hr. Of note, the NRVMs were placed in the bottom of a live imaging dish and the hydrogel was added to the outer ledge, not in direct contact with the cells.
2.11. Western blots
NRVMs were rinsed 2x with cold 1x Tris-buffered saline (TBS) and then homogenized in lysis buffer (50 mM Tris-HCl [pH 7.4], 150 mM NaCl, 0.2% SDS, Complete protease inhibitor, and PhosSTOP) on ice for 30 min14. Protein concentration was quantified by BCA assay (Pierce). A total of 30 μg protein lysate was resolved by SDS-PAGE and transferred to PVDF (EMD Millipore) membrane. Blots were blocked in 5% BSA in TBST (1x TBS, 0.1% Tween 20) for 1 hr at RT and incubated with indicated primary antibody in blocking buffer overnight at 4°C. Blots were washed 3× 10 min with TBST, and then incubated for 1 hr at RT with secondary antibody. Blots were then washed again 3× 10 min with TBST, detected using the SuperSignal West Femto Maximum Sensitivity (ThermoFisher) per manufacturer protocol, and imaged with an Invitrogen iBright Imager. Quantifications were done using NIH ImageJ software using a minimum of three independent replicates.
The following antibodies were used in this study: the α-phospho-Cx43 (Y247) antibody was custom produced in rabbit from a peptide designed and chemically synthesized by LifeTein. Additional antibodies used in this study include α-Src (#2123S), α-phospho-Src (Y416) (#6943S), α- protein kinase C (PKC) (#2056), α-phospho-PKC (S660) (#9375), α-Erk1/2 (#4695), α-phospho-Erk1/2 (T202/Y204) (#4370), α-JNK (#9252), α-phospho-JNK (T183/Y185) (#4668), α-Pyk2 (#3480S), α-Cleaved Caspase 9 (#9505S), α-rabbit-Alexa488 (#4412S), α-mouse-Alexa647 (#4410S), α-rabbit-HRP (#7074), and α-mouse-HRP (#7076) purchased from Cell Signaling; α-phospho-Pyk2 (Y579/580) (#44–636G) purchased from ThermoFisher; ANP (sc-515701) purchased from Santa Cruz; α-Cx43 (#C6219) purchased from Sigma; α-phospho-Cx43 (Y265) (ab193373), α-phospho-Pyk2 (Y402) (ab4800), and α-alpha actinin (ab90421) purchased from Abcam. DAPI (#5748) was purchased from Tocris Bioscience.
2.12. TUNEL Assay
The apoptosis of NRVMs was measured using TUNEL Assay Kit (Roche) per manufacturer protocol (catalog number: 11684817910)51. Primary cardiomyocytes were pre-seeded on live imaging dishes coated with Fibronectin. Cells were fixed with 4% PFA for 1 hr and then rinsed with PBS and incubated with permeabilization solution (0.1% Triton X-100 in 0.1% sodium citrate) for 2 min on ice. Then the cells were rinsed with PBS and incubated with TUNEL reaction mixture at 37°C for 60 min, followed by incubation with DAPI for 10 min. The results were observed and quantified by confocal fluorescence microscopy.
2.13. Immunofluorescence
NRVMs were pre-seeded on coverslips coated with Fibronectin. Cells were fixed with 4% PFA for 30 min and then washed with TBST and blocked (1x TBS with 2.5% horse serum and 0.2% Triton) for 1 hr at RT. Cells on the coverslips were incubated at 4°C overnight with primary antibodies. The next day, coverslips were washed 3× 10 min with 1x TBST, incubated with secondary antibodies for 1 hr at RT, stained with DAPI (100 ng/mL) for 10 min, and then washed 3× 10 min with 1x TBST. Coverslips were mounted on a drop of SlowFade anti-fade (Life Tech), sealed with clear nail polish, and imaged.
2.14. Confocal imaging
All cell immunofluorescence images were acquired on a Zeiss LSM 800 Confocal system using appropriate numerical aperture objectives and appropriate filter sets.
2.15. Statistical Methods
All data were analyzed by using GraphPad Prism 8.0 and presented as the mean ± SD. Statistical analysis performed in GraphPad Prism 8.0 were either one-way ANOVA with a Neuman-Keuls post-hoc analysis or Student’s t-test where appropriate. P-values <0.05 were considered statistically significant.
3. Results
3.1. Synthesis and characterization of HA-PM hydrogels.
A micelle crosslinked hydrogel system was developed to deliver two TKIs simultaneously to the NVCMs, thereby to synergistically influence Cx43 gap junction intercellular communication (Fig. 1). Two polymer precursors were first synthesized before the fabrication of the hydrogels.
The HA was conjugated with the cysteamine by EDC/NHS mediated coupling and 1H-NMR result revealed a roughly 20% grafting ratio (Fig. 2A). The hydroxyl terminal of the Pluronic P123 was first activated by the succinic anhydride at high efficiency (~90% determined by 1H-NMR). The P123 dicarboxylate was further coupled with the N-(2-aminoethyl) maleimide and the resulting PM showed a ~45% grafting ratio (Fig. 2B).
Figure 2. NMR characterization of synthesized hydrogel precursors.
A) 1H NMR spectra of HA and HA-SH. B) 1H NMR spectra of P123, P123-succinic acid, and P123-maleimide.
PM was dissolved in water at 5% and HA-SH was prepared at two concentrations: 1% and 2%, in water, respectively. The hydrogels were fabricated by mixing the two polymer conjugates together using the thiol-ene click reaction. Both 1% and 2% HA-SH formed hydrogels with the 5% PM rapidly (Fig. 3A). The time sweep testing indicated the storage modulus (G’) of both hydrogels plateaued within 8 min at 37°C. The 5:1 hydrogel showed a G’ of 140 ± 10 Pa and loss modulus (G”) of 15 ± 5 Pa while the 5:2 hydrogel showed a G’ of 355 ± 25 Pa and G” of 30 ± 10 Pa. Both hydrogels maintained their elastic behavior over the tested frequency range (0.1 – 10 Hz) as shown by the continual G’ > G” (Fig. 3B). FTIR further confirmed the two major polymer components in the hydrogel (Supplemental Fig. 1). The hydrogels possessed shear-thinning ability as confirmed by the sharp viscosity decrease when the shear rate ramped from 1 s−1 to 100 s−1 (Fig. 3C). The viscosity of the 5:2 hydrogel decreased sharply from 160 Pa-s at 1 s−1 shear rate to 10 Pa-s at 100 s−1. Meanwhile, the viscosity of the 5:1 hydrogel dropped from 74 Pa-s at 1 s−1 to 1.3 Pa-s at 100 s−1. The hydrogels were injectable and could be smoothly extruded through a 29 G needle (Supplemental Video 1). The compressive stress of both hydrogels was also characterized, and the recovery study also proved good injectability of the hydrogels (Supplemental Fig. 2).
Figure 3. Characterization of injectable HA-PM hydrogels.
A) Time sweep rheological testing indicating the storage modulus (G’) and loss modulus (G”) change of two hydrogels with different precursor ratio over time. (B) Frequency sweep rheological. (C) Viscosity of the two hydrogels with shear rate increasing from 1 s−1 to 100 s−1. The inserted image indicated that a dye-colored hydrogel was smoothly injectable through a 29 G needle. D) Swelling ratio of the two hydrogels over 24 hr. E) Degradation profile of two hydrogels over 21 days. F) Cyto-compatibility of the hydrogels determined by MTT study (n=4; **, p<0.01).
The 5:2 hydrogel showed more swelling ratio compared to the 5:1 hydrogel. This was probably caused by the higher amount of hydrophilic HA component in the 5:2 hydrogel (Fig. 3D). The 5:2 hydrogel was also relatively more stable than the 5:1 hydrogel in PBS (Fig. 3E). The 5:1 hydrogel degraded ~65% in 21 days while the 5:2 hydrogel only degraded ~25% in the same period. Finally, the cyto-compatibility of the hydrogels was evaluated on the NVCMs and the result implied no obvious cytotoxicity on the cells for the 5:2 hydrogel but nearly 15% decrease for the 5:1 hydrogel based on the MTT study (Fig. 3F). We decided to use the 5:2 hydrogel for further studies due to its better stability and cytocompatibility.
3.2. In vitro drug release from the TKIs loaded hydrogels.
We found Pluronic P123 could effectively encapsulate two TKIs, i.e., Saracatinib and PF4618433, presumably due to the micelle formation. On the other hand, Pluronic F-127 was able to encapsulate Saracatinib but not PF4618433, making it unsuitable for our purposes. In the 5% PM solution, we were able to dissolve up to 4 mg/mL of Saracatinib and 2 mg/mL of PF4618433, achieving an encapsulation efficiency of over 95%. However, our previous study indicated Saracatinib at 4 mg/mL was just as effective as 0.2 mg/mL to improve Cx43 gap junction intercellular communication14. Therefore, we prepared the drug loaded PM micelles with 0.2 mg/mL Saracatinib and 2 mg/mL PF4618433. We compared the particle size and PdI of micelles loaded with different drugs using dynamic light scattering. The results showed that PF4618433 loaded micelles had comparable size with the Saracatinib loaded micelles, although the PdI of the PF4618433 micelles was significantly smaller than the Saracatinib micelles (Fig. 4A, B). This was probably because the PF4618433 was more hydrophobic than the Saracatinib and could be packed more tightly in the hydrophobic cores of the micelles. When both drugs were loaded, the dual drug loaded micelles showed similar size and PdI compared to the PF4618433 loaded micelles. The encapsulated drugs did not have adverse effect in the hydrogel formation.
Figure 4. Characterization of drug loaded micelles and drug release from the micelles and hydrogels.
A, B) Dynamic light scattering analysis of different drug loaded micelles. C) SEM image of HA-PM hydrogel loaded with both drugs. D) Representative HPLC chromatography of dual-drug separation under the optimized gradient mobile phase condition. E) Dual drug release profile from the hydrogels over a 14 day period. SA, Saracatinib; PF, PF4618433.
The G’ and G” of the dual drug loaded micelle crosslinked hydrogels were comparable to the empty micelle crosslinked hydrogels. Noteworthy, the 5:2 hydrogel showed relatively smaller pores compared to the 5:1 hydrogel due to increased crosslinking degree (Fig. 4C). No obvious drug precipitates/aggregates were found in the SEM image of the hydrogels, indicating successful encapsulation and dispersion of the drugs in the micelles and hydrogels. This is also proved by the FTIR spectra of dual drug loaded hydrogel, which showed the characteristic peaks of dual drugs, validating the presence of both drugs in the hydrogel (Supplemental Fig. 3). HPLC was used to monitor the drug release, which could determine the concentrations of both drugs at the same time from the releasate (Fig. 4D). Saracatinib was released faster from the hydrogels than PF4618433 because it is more hydrophilic. It showed a burst release of around 43% on day 1 and an average of 92% release on day 14 (Fig. 4E). In contrast, there was only 12% of PF4618433 released on day 1 and an average of 32% was released on day 14.
3.3. Hydrogel release of the Pyk2 inhibitor PF4618433 has the same effect on PMA treated NRVMs as when directly added to the media.
Due to the lower release from the hydrogel of PF4618433 when compared to Saracatinib, here we initially tested if the biological effect of the PF4618433 would be similar to when the inhibitor was directly added to the media14. NRVMs were pretreated with PF4618433 and then exposed to PMA, which we and others have shown activates Pyk2 and Src leading to increased tyrosine phosphorylation of Cx43 as well as down regulation of gap junction intercellular communication14, 52. Western blot data demonstrates that PF4618433 directly added to the media or released from the hydrogels reduced the activation of Src and Pyk2 in a similar manner (Fig. 5A, B). The level of phosphorylation on Cx43 residues Y247 and Y265 was also equally reduced by both delivery methods of PF4618433. The data suggests PF4618433 released from the hydrogel can influence signaling pathways and Cx43 regulation in NRVMs.
Figure 5. Comparing the effect of PF4618433 (PF) when released from the hydrogel to when directly added to the media on tyrosine kinase activity and Cx43 regulation.
A) NRVMs were pre-treated with PF (5 μM) in medium or in hydrogel (10 μg) for 3 hr and then treated with PMA (300 nM, 60 min). Lysate from each group was Western blotted. Antibodies used are labeled on the left of each panel. B) Protein levels (PF in medium, top; PF in hydrogel, below) were quantified by analyzing scanned blots using ImageJ software Phosphorylated protein levels were normalized to total protein. Data are representative of three independent experiments (one-way ANOVA, n=3, *P < 0.05, **P < 0.01, ***P < 0.001).
3.4. Hydrogel release of the Src inhibitor Saracatinib or Pyk2 inhibitor PF4618433 maintains Cx43 at the plasma membrane in PMA treated NRVMs.
As a control, presented is immunofluorescence data demonstrating that 60 min of PMA treatment causes internalization of Cx43 from the plasma membrane (Fig. 6A). Sarcomeric α-actinin was used as a cardiac cell marker. Release of the Src inhibitor Saracatinib from the hydrogel increased the level of plasma membrane Cx43, however a significant amount still remained intracellular. The Pyk2 inhibitor PF4618433 was more effective than Saracatinib at maintaining Cx43 at the plasma membrane. The combination of Saracatinib and PF4618433 (Dual) had no additional effect than PF4618433 alone. These observations in NRVMs were identical when both TKIs were directly added to the media prior to the addition of PMA14.
Figure 6. Effect of Saracatinib and PF4618433 (PF) release from the hydrogel on cellular localization of Cx43.
A) NRVMs cultured in 12-well plate were pre-treated with hydrogel containing Saracatinib (1 μg/well), PF4618433 (10 μg/well), or both (Dual, 3 h) and then treated with PMA (300 nM, 60 min). A representative fluorescent image is shown for each group (red, cardiac marker sarcomeric α-actinin; green, Cx43; blue, DAPI; white arrows, Cx43 in the junctional fraction at the plasma membrane; yellow arrows, internalized Cx43). B) Quantification was the percentage of total fluorescent intensity of Cx43 at GJ plaque to total Cx43. (one-way ANOVA, *P < 0.05, ***P < 0.001).
3.5. Hydrogel release of the Src inhibitor Saracatinib or Pyk2 inhibitor PF4618433 reverses PMA induced hypertrophy in NVCMs.
PMA activates the fetal gene program (e.g., ANP) and increases cardiomyocyte size via PKC signaling41, 42. Here we investigated if Saracatinib and/or PF4618433 could reverse the hypertrophic response to PMA. Immunofluorescence data demonstrates that 24 hr of PMA treatment causes an increase in ANP expression and cell area (Fig. 7A, B). The release of Saracatinib or PF4618433 from the hydrogels had a similar effect to decrease the level of cells expressing ANP induced by PMA treatment back to the level of control no PMA. Interestingly, when both inhibitors were released together (Dual), they statistically had a lower level of ANP than either Saracatinib or PF4618433 alone (albeit still not different from control no PMA). The release of Saracatinib or PF4618433 had a similar effect to decrease the cell area compared to PMA treatment group, with the combination of both have no additive effect.
Figure 7. Effect of Saracatinib and PF4618433 (PF) release from the hydrogel on PMA mediated hypertrophy.
A) NRVMs cultured in 12-well plate were pre-treated with Saracatinib (SA, 1 μg/well), PF4618433 (PF, 10 μg/well), or both (Dual, 3 hr) and then treated with PMA (300 nM, 24 hr). A representative fluorescent image is shown for each group (red, cardiac marker sarcomeric α-actinin; green, ANP; blue, DAPI). B) Cell surface area and percentage of total fluorescent intensity of ANP to DAPI were quantified using Image J software. C) Lysate from each group was Western blotted. Antibodies used are labeled on the left of each panel. D) Protein levels were quantified by analyzing scanned blots using Image J software, with normalization of protein expression to the control lane (value set arbitrarily as 1). Phosphorylated protein levels were normalized to total protein. Data are representative of three independent experiments (one-way ANOVA, n=3, *P < 0.05, **P < 0.01, ***P < 0.001).
Next, signaling pathways activated by PMA were evaluated to see which may have been downregulated to contribute to the decreased ANP level and cell size. Western blot data demonstrates that 24 hr of PMA treatment activates PKC, ERK, and JNK (Fig. 7C, D). The release of Saracatinib from the hydrogel decreased the activity of PKC (but not to control no PMA level) and had no effect on ERK and JNK. Conversely, PF4618433 decreased the activity of PKC, ERK, and JNK, with PKC and ERK returning to the control no PMA level. The combination of both Saracatinib and PF4618433 (Dual) had no additional effect on PKC, ERK, and JNK activity than PF4618433 alone.
3.6. Hydrogel release of the Src inhibitor Saracatinib or Pyk2 inhibitor PF4618433 reverses ET-1 and phenylephrine induced hypertrophy and apoptosis of NVCMs.
ET-1, a vasoconstrictor peptide secreted from vascular endothelial cells as well as by cardiomyocytes, induces hypertrophy of cardiomyocytes through activating phospholipase C, PKC, Erk1/Erk2, and upregulation of c-Fos, c-Jun, and JNK53, 54. Immunofluorescence data demonstrates that 24 hr of ET-1 treatment causes an increase in ANP and cell area (Fig. 8A, B). The release of Saracatinib or PF4618433 from the hydrogels had a similar effect to decrease the level of cells expressing ANP back to the level of control no ET-1. Like the treatment with PMA, when both inhibitors were released together (Dual), they statistically had a lower level of ANP than either Saracatinib or PF4618433 alone. The release of either Saracatinib or PF4618433 had a similar effect to decrease the cell area. However, when both inhibitors were combined (Dual), there was no enhanced effect as was seen for ANP. Western blot data demonstrates that 24 hr of ET-1 treatment activates Src, Pyk2, PKC, ERK, and JNK (Fig. 8C, D). Release of Saracatinib or PF4618433 form the hydrogels reduced the activity of Src, Pyk2, PKC, JNK and ERK all back to the control level. The combination of Saracatinib and PF4618433 (Dual) had no additional effect than PF4618433 alone.
Figure 8. Effect of Saracatinib and PF4618433 release from the hydrogel on ET-1 mediated hypertrophy.
A) NRVMs cultured in 12-well plate were pre-treated with Saracatinib (SA, 1 μg/well), PF4618433 (PF, 10 μg/well), or both (Dual, 3 hr) and then treated with ET-1 (100 nM, 24 hr). A representative fluorescent image is shown for each group (red, cardiac marker sarcomeric α-actinin; green, ANP; blue, DAPI). B) Cell surface area and percentage of total fluorescent intensity of ANP to DAPI were quantified using Image J software. C) Lysate from each group was Western blotted. Antibodies used are labeled on the left of each panel. D) Protein levels were quantified by analyzing scanned blots using Image J software, with normalization of protein expression to the control lane (value set arbitrarily as 1). Phosphorylated protein levels were normalized to total protein. Data are representative of three independent experiments. E) The apoptosis of NRVMs was detected using TUNEL assay. F) The TUNEL-positive cells were quantified using Image J software. Data are representative of three independent experiments (one-way ANOVA, n=3, *P < 0.05, **P < 0.01, ***P < 0.001).
The TUNEL assay detects DNA breakage by labeling the free 3ʹ-hydroxyl termini. Considering that genomic DNA breaks occur during apoptosis, we utilized TUNEL staining as a means to quantify apoptotic cell death induced by ET-1. Immunofluorescence results demonstrate that 24 hr of ET-1 treatment causes an increase in NRVMs undergoing apoptosis (Fig. 8E). Release of Saracatinib or PF4618433 from the hydrogels decreased the level of apoptosis, however not back to the control level. The combination of Saracatinib and PF4618433 (Dual) had a greater effect than either inhibitor alone as the amount of apoptosis returned to the control level without ET-1.
Phenylephrine, a selective α1-adrenergic receptor (α1-AR) agonist, is widely used as a stimulus for hypertrophy in cardiomyocytes by activating a number of the same signalling pathways as ET-155, 56. Using the TUNEL assay, immunofluorescence data demonstrates that 24 hr of phenylephrine treatment causes an increase in NRVMs undergoing apoptosis (Fig. 9A, B).
Figure 9. Effect of Saracatinib and PF4618433 release from the hydrogels on phenylephrine mediated hypertrophy.
A) NRVMs cultured in 12-well plate were pre-treated with Saracatinib (SA, 1 μg/well), PF4618433 (PF, 10 μg/well), or both (Dual, 3 hr) and then treated with phenylephrine (100 nM, 24 hr). The apoptosis of NRVMs was detected using TUNEL assay. The TUNEL-positive cells were quantified using Image J software (B). C) A representative fluorescence image of apoptosis-related protein (cleaved caspase 9) is shown for each group (red, cardiac marker sarcomeric α-actinin; green, cleaved caspase 9; blue, DAPI). D) Percentage of total fluorescence intensity of cleaved caspase 9 to DAPI was quantified using Image J software. (one-way ANOVA, *P < 0.05, **P < 0.01, ***P < 0.001).
Release of Saracatinib or PF4618433 from the hydrogels decreased the level of apoptosis, however the level did not return to the control level without phenylephrine. The combination of Saracatinib and PF4618433 (Dual) had a stronger effect than either inhibitor alone as the amount of apoptosis returned to the control level without phenylephrine.
To validate the apoptosis results, we conducted an additional test using another apoptosis marker, Caspase-9, which belongs to the cysteine aspartic acid protease (caspase) family57. By employing an antibody that recognized activated Caspase-9 (cleaved version), we observed similar results in the immunofluorescence data as those obtained from the TUNEL assay. The release of Saracatinib or PF4618433 from the hydrogels resulted in a reduction in the level of apoptosis. However, this reduction did not reach the level observed in the control group without phenylephrine. The combined use of Saracatinib and PF4618433 (Dual) exhibited a stronger effect compared to either inhibitor used individually, returned to the control level without phenylephrine.
4. Discussion
MI is a leading cause of death globally and loss of cardiomyocytes plays a critical role in the pathogenesis of heart failure58. With fewer cardiomyocytes, the heart is unable to sustain efficient contraction. Cardiomyocyte death leads to a series of pathological remodeling through the accumulation of fibrous tissue59. The lack of electrical connection between the healthy myocardium and islands of intact cardiomyocytes in fibrotic matrices support asynchronous ventricular contraction, resulting in progressive functional decompensation60. Several current heart failure therapies aim to promote cardiac repair by modifying immune cell infiltration and extracellular matrix composition, promoting angiogenesis and heart regeneration, and reducing cardiac hypertrophy61–65.
An additional therapeutic to improve cardiac function would be to decrease the initial cardiomyocyte cell death in order to maintain proper impulse propagation. Implicated in this process is Cx43, as within 1 hour post-MI, expression is reduced and localization becomes lateralized at the infarct border zone; the same phenotype occurs in later in the disease during left ventricle hypertrophy8, 66 67. Cx43 has been shown to be a promising therapeutic target in cardiovascular disease66, 68, 69. For example, the peptide αCT1, which mimics the Cx43 residues involved in binding the ZO-1 PDZ2 domain, has anti-arrhythmic effects, as well as cardioprotective properties in mouse hearts subject to ischemia reperfusion injuries70, 71. Inhibition of Pyk2 activity using the small molecule compound, PF4618433, reduced infarct size, maintained Cx43 at the intercalated disc in left ventricle hypertrophic myocytes, and improved cardiac function in an animal model26. The Src inhibitor Saracatinib, raised Cx43 expression, improved conduction velocity, and lowered arrhythmia inducibility13. In this study, we identified mechanisms by which PF4618433 and Saracatinib elicit these positive responses post-MI. After PMA or ET-1 stimulation, PF4618433 and Saracatinib decreased expression of ANP. Plasma levels of ANP (and brain natriuretic peptide) and are elevated immediately post-MI as well as in heart failure and are predictors of ventricular dysfunction and mortality72–74. We speculate that PF4618433 and Saracatinib are influencing the transcription factor GATA4, which is involved in ANP gene expression75. This observation is consistent with the decreased cell area observed in the presence of PF4618433 and Saracatinib as ANP is a biomarker for hypertrophy76.
PF4618433 had a greater effect than Saracatinib to decrease the activation of PKC, ERK, and JNK after PMA treatment, while these TKIs had a similar decreased effect decreases after ET-1 treatment. A number of studies have placed PKC activation upstream of Pyk2 activation77, 78. However, one study identified that Pyk2 and PKCα can mutually influence each other to regulating Nanog and Oct4 transcription factors and the stemness phenotype in triple negative breast cancer79. Conversely, Pyk2 is upstream of JNK and ERK activation and mediates its response through the adaptor proteins Crk and Grb2, respectively80. Similar to Pyk2, Src directly activates PKCδ in many models of cardiac ischemia and hypertrophy and is upstream of JNK and ERK activation81–83. Observations from the treatment with phenylephrine were similar to that of ET-1 (and PMA), in that there was an increase in apoptosis and cell area. These were reversed in the presence of PF4618433 or Saracatinib. The presence of both PF4618433 and Saracatinib further decreased the level of ANP and apoptosis than each inhibitor alone, suggesting a combinatorial approach may be most beneficial.
Based on these characteristics, PF4618433 and Saracatinib hold promise as potential therapeutics for enhancing cardiac function in humans. In support of this, we have presented evidence that Pyk2 is activated following MI in the human left ventricle26. While PF4618433 was proven safe in an animal model of MI induced heart failure, PF4618433 was applied via intraperitoneally injection26. Unfortunately, this route of administration may not be optimal for directly targeting the left ventricle at the necessary concentrations during the acute phase post-MI to prevent cardiomyocyte death, nor for the continuous application required over an extended period to support the hypertrophic heart26. An injectable hydrogel-based delivery system holds promise as a more viable option. These hydrogels have been specifically designed for cardiac repair, with a focus on restoring electrical signaling propagation across the infarct region36, 84. These hydrogels offer multiple functionalities, including mechanical support, improvement of the extracellular matrix, enhanced conductivity, scavenging of reactive oxygen species, and reduction in the expression of inflammatory cytokines35, 85, 86. Here we provide the first step towards the proof of concept that injectable hydrogels, consisting of HA-SH and PM micelles, could also be used to release kinase inhibitors to decrease cell death by promoting intercellular communication. PM micelles can effectively incorporate both PF4618433 and Saracatinib and achieve sustained release, which cannot be accomplished by using Pluronic F-127 micelles. In addition, PM micelles act as a crosslinking agent for HA-SH, facilitating the formation of injectable hydrogels with shear-thinning properties. This hydrogel system can be delivered locally to the myocardium through transcatheters, allowing for targeted delivery of the dual TKIs. The next step in this study will be to revisit the MI model of heart failure were the hydrogel containing PF4618433 and Saracatinib is applied to the myocardium supplied by the left anterior descending artery and distal to this region that undergoes hypertrophy.
6. Conclusions
We developed an injectable hydrogel consisting of HA-SH and PM micelles, that can be injectable, incorporate PF4618433 and Saracatinib, and achieve sustained release. The significance of this study is that PF4618433 had the same effect released from the hydrogel as directly added to the media to reduce Src and Pyk2 activity as well as reduce tyrosine phosphorylation of Cx43 residues associated with a decrease in gap junction intercellular communication. Additionally, the presence of both PF4618433 and Saracatinib further decreased the level of ANP and apoptosis than each inhibitor alone, suggesting a combinatorial approach may be most beneficial. This delivery system holds promising for further intracardiac, pericardial or epicardial injection to treat MI.
Supplementary Material
Acknowledgements
We would like to thank the Advanced Microscopy Core Facility at the University of Nebraska Medical Center for helping with confocal microscopy.
Sources of Funding
This work was supported by a grant from the NIH (GM072631) to P. L. Sorgen. Support for the University of Nebraska Medical Center Advanced Microscopy Core Facility was provided by the Nebraska Research Initiative and an Institutional Development Award from the NIH (P30GM106397). The following NIH SIG funded instruments were used: LSM 800 Zeiss Confocal Microscope (NIH S10RR027301).
Reference
- (1).Krijnen PA; Nijmeijer R; Meijer CJ; Visser CA; Hack CE; Niessen HW Apoptosis in myocardial ischaemia and infarction. J Clin Pathol 2002, 55 (11), 801–811. DOI: 10.1136/jcp.55.11.801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (2).Jeyaraman M; Tanguy S; Fandrich RR; Lukas A; Kardami E Ischemia-induced dephosphorylation of cardiomyocyte connexin-43 is reduced by okadaic acid and calyculin A but not fostriecin. Mol Cell Biochem 2003, 242 (1–2), 129–134. [PubMed] [Google Scholar]
- (3).Turner MS; Haywood GA; Andreka P; You L; Martin PE; Evans WH; Webster KA; Bishopric NH Reversible connexin 43 dephosphorylation during hypoxia and reoxygenation is linked to cellular ATP levels. Circ Res 2004, 95 (7), 726–733. DOI: 10.1161/01.RES.0000144805.11519.1e. [DOI] [PubMed] [Google Scholar]
- (4).Matsumura K; Mayama T; Lin H; Sakamoto Y; Ogawa K; Imanaga I Effects of cyclic AMP on the function of the cardiac gap junction during hypoxia. Exp Clin Cardiol 2006, 11 (4), 286–293. [PMC free article] [PubMed] [Google Scholar]
- (5).Lampe PD; Cooper CD; King TJ; Burt JM Analysis of Connexin43 phosphorylated at S325, S328 and S330 in normoxic and ischemic heart. J Cell Sci 2006, 119 (Pt 16), 3435–3442. DOI: 10.1242/jcs.03089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (6).Kleber AG; Rudy Y Basic mechanisms of cardiac impulse propagation and associated arrhythmias. Physiol Rev 2004, 84 (2), 431–488. DOI: 10.1152/physrev.00025.2003. [DOI] [PubMed] [Google Scholar]
- (7).Kieken F; Mutsaers N; Dolmatova E; Virgil K; Wit AL; Kellezi A; Hirst-Jensen BJ; Duffy HS; Sorgen PL Structural and molecular mechanisms of gap junction remodeling in epicardial border zone myocytes following myocardial infarction. Circ Res 2009, 104 (9), 1103–1112. DOI: 10.1161/CIRCRESAHA.108.190454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (8).Peters NS; Coromilas J; Severs NJ; Wit AL Disturbed connexin43 gap junction distribution correlates with the location of reentrant circuits in the epicardial border zone of healing canine infarcts that cause ventricular tachycardia. Circulation 1997, 95 (4), 988–996. DOI: 10.1161/01.cir.95.4.988. [DOI] [PubMed] [Google Scholar]
- (9).Frey N; Katus HA; Olson EN; Hill JA Hypertrophy of the heart: a new therapeutic target? Circulation 2004, 109 (13), 1580–1589. DOI: 10.1161/01.CIR.0000120390.68287.BB. [DOI] [PubMed] [Google Scholar]
- (10).Jin H; Chemaly ER; Lee A; Kho C; Hadri L; Hajjar RJ; Akar FG Mechanoelectrical remodeling and arrhythmias during progression of hypertrophy. FASEB J 2010, 24 (2), 451–463. DOI: 10.1096/fj.09-136622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (11).Shiojima I; Sato K; Izumiya Y; Schiekofer S; Ito M; Liao R; Colucci WS; Walsh K Disruption of coordinated cardiac hypertrophy and angiogenesis contributes to the transition to heart failure. J Clin Invest 2005, 115 (8), 2108–2118. DOI: 10.1172/JCI24682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (12).Cooklin M; Wallis WR; Sheridan DJ; Fry CH Changes in cell-to-cell electrical coupling associated with left ventricular hypertrophy. Circ Res 1997, 80 (6), 765–771. DOI: 10.1161/01.res.80.6.765. [DOI] [PubMed] [Google Scholar]
- (13).Rutledge CA; Ng FS; Sulkin MS; Greener ID; Sergeyenko AM; Liu H; Gemel J; Beyer EC; Sovari AA; Efimov IR; et al. c-Src kinase inhibition reduces arrhythmia inducibility and connexin43 dysregulation after myocardial infarction. J Am Coll Cardiol 2014, 63 (9), 928–934. DOI: 10.1016/j.jacc.2013.10.081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (14).Zheng L; Trease AJ; Katsurada K; Spagnol G; Li H; Shi W; Duan B; Patel KP; Sorgen PL Inhibition of Pyk2 and Src activity improves Cx43 gap junction intercellular communication. J Mol Cell Cardiol 2020, 149, 27–40. DOI: 10.1016/j.yjmcc.2020.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (15).Akar FG; Nass RD; Hahn S; Cingolani E; Shah M; Hesketh GG; DiSilvestre D; Tunin RS; Kass DA; Tomaselli GF Dynamic changes in conduction velocity and gap junction properties during development of pacing-induced heart failure. Am J Physiol Heart Circ Physiol 2007, 293 (2), H1223–1230. DOI: 10.1152/ajpheart.00079.2007. [DOI] [PubMed] [Google Scholar]
- (16).Kaplan SR; Gard JJ; Carvajal-Huerta L; Ruiz-Cabezas JC; Thiene G; Saffitz JE Structural and molecular pathology of the heart in Carvajal syndrome. Cardiovasc Pathol 2004, 13 (1), 26–32. [DOI] [PubMed] [Google Scholar]
- (17).Saidi Brikci-Nigassa A; Clement MJ; Ha-Duong T; Adjadj E; Ziani L; Pastre D; Curmi PA; Savarin P Phosphorylation controls the interaction of the connexin43 C-terminal domain with tubulin and microtubules. Biochemistry 2012, 51 (21), 4331–4342. DOI: 10.1021/bi201806j. [DOI] [PubMed] [Google Scholar]
- (18).Zheng L; Li H; Cannon A; Trease AJ; Spagnol G; Zheng H; Radio S; Patel K; Batra S; Sorgen PL Phosphorylation of Cx43 residue Y313 by Src contributes to blocking the interaction with Drebrin and disassembling gap junctions. J Mol Cell Cardiol 2019, 126, 36–49. DOI: 10.1016/j.yjmcc.2018.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (19).Ambrosi C; Ren C; Spagnol G; Cavin G; Cone A; Grintsevich EE; Sosinsky GE; Sorgen PL Connexin43 Forms Supramolecular Complexes through Non-Overlapping Binding Sites for Drebrin, Tubulin, and ZO-1. PLoS One 2016, 11 (6), e0157073. DOI: 10.1371/journal.pone.0157073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (20).Hunter AW; Barker RJ; Zhu C; Gourdie RG Zonula occludens-1 alters connexin43 gap junction size and organization by influencing channel accretion. Mol Biol Cell 2005, 16 (12), 5686–5698. DOI: 10.1091/mbc.e05-08-0737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (21).Dunn CA; Lampe PD Injury-triggered Akt phosphorylation of Cx43: a ZO-1-driven molecular switch that regulates gap junction size. J Cell Sci 2014, 127 (Pt 2), 455–464. DOI: 10.1242/jcs.142497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (22).Cabo C; Yao J; Boyden PA; Chen S; Hussain W; Duffy HS; Ciaccio EJ; Peters NS; Wit AL Heterogeneous gap junction remodeling in reentrant circuits in the epicardial border zone of the healing canine infarct. Cardiovasc Res 2006, 72 (2), 241–249. DOI: 10.1016/j.cardiores.2006.07.005. [DOI] [PubMed] [Google Scholar]
- (23).Cabo C; Boyden PA Extracellular space attenuates the effect of gap junctional remodeling on wave propagation: a computational study. Biophys J 2009, 96 (8), 3092–3101. DOI: 10.1016/j.bpj.2009.01.014 From NLM Medline. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (24).Ek-Vitorin JF; King TJ; Heyman NS; Lampe PD; Burt JM Selectivity of connexin 43 channels is regulated through protein kinase C-dependent phosphorylation. Circ Res 2006, 98 (12), 1498–1505. DOI: 10.1161/01.RES.0000227572.45891.2c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (25).Macia E; Dolmatova E; Cabo C; Sosinsky AZ; Dun W; Coromilas J; Ciaccio EJ; Boyden PA; Wit AL; Duffy HS Characterization of gap junction remodeling in epicardial border zone of healing canine infarcts and electrophysiological effects of partial reversal by rotigaptide. Circ Arrhythm Electrophysiol 2011, 4 (3), 344–351. DOI: 10.1161/CIRCEP.110.959312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (26).Zheng L; Spagnol G; Gandhi DR; Sharma K; Kumar V; Patel KP; Sorgen PL Inhibition of Pyk2 Improves Cx43 Intercalated Disc Localization, Infarct Size, and Cardiac Function in Rats With Heart Failure. Circ Heart Fail 2023, e010294. DOI: 10.1161/CIRCHEARTFAILURE.122.010294 From NLM Publisher. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (27).Han S; Mistry A; Chang JS; Cunningham D; Griffor M; Bonnette PC; Wang H; Chrunyk BA; Aspnes GE; Walker DP; et al. Structural characterization of proline-rich tyrosine kinase 2 (PYK2) reveals a unique (DFG-out) conformation and enables inhibitor design. J Biol Chem 2009, 284 (19), 13193–13201. DOI: 10.1074/jbc.M809038200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (28).Gil-Cabrerizo P; Scacchetti I; Garbayo E; Blanco-Prieto MJ Cardiac tissue engineering for myocardial infarction treatment. Eur J Pharm Sci 2023, 185, 106439. DOI: 10.1016/j.ejps.2023.106439 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (29).Hu W; Yang C; Guo X; Wu Y; Loh XJ; Li Z; Wu YL; Wu C Research Advances of Injectable Functional Hydrogel Materials in the Treatment of Myocardial Infarction. Gels 2022, 8 (7). DOI: 10.3390/gels8070423 From NLM PubMed-not-MEDLINE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (30).Lu Y; Luo Y; Zhu R; Huang X; Bai S Advanced bioactive hydrogels for the treatment of myocardial infarction. J Mater Chem B 2022, 10 (41), 8375–8385. DOI: 10.1039/d2tb01591a From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (31).Zhu D; Li Z; Huang K; Caranasos TG; Rossi JS; Cheng K Minimally invasive delivery of therapeutic agents by hydrogel injection into the pericardial cavity for cardiac repair. Nature communications 2021, 12 (1), 1412. DOI: 10.1038/s41467-021-21682-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (32).Guvendiren M; Burdick JA Stiffening hydrogels to probe short- and long-term cellular responses to dynamic mechanics. Nature communications 2012, 3, 792. DOI: 10.1038/ncomms1792. [DOI] [PubMed] [Google Scholar]
- (33).Seif-Naraghi SB; Singelyn JM; Salvatore MA; Osborn KG; Wang JJ; Sampat U; Kwan OL; Strachan GM; Wong J; Schup-Magoffin PJ; et al. Safety and efficacy of an injectable extracellular matrix hydrogel for treating myocardial infarction. Sci Transl Med 2013, 5 (173), 173ra125. DOI: 10.1126/scitranslmed.3005503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (34).Rodell CB; Lee ME; Wang H; Takebayashi S; Takayama T; Kawamura T; Arkles JS; Dusaj NN; Dorsey SM; Witschey WR; et al. Injectable Shear-Thinning Hydrogels for Minimally Invasive Delivery to Infarcted Myocardium to Limit Left Ventricular Remodeling. Circ Cardiovasc Interv 2016, 9 (10). DOI: 10.1161/CIRCINTERVENTIONS.116.004058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (35).Wu T; Liu W Functional hydrogels for the treatment of myocardial infarction. ngp asia materials 2022, 14 (9), Review. [Google Scholar]
- (36).Zhang L; Li T; Yu Y; Shi K; Bei Z; Qian Y; Qian Z An injectable conductive hydrogel restores electrical transmission at myocardial infarct site to preserve cardiac function and enhance repair. Bioact Mater 2023, 20, 339–354. DOI: 10.1016/j.bioactmat.2022.06.001 From NLM PubMed-not-MEDLINE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (37).Ma XL; Kumar S; Gao F; Louden CS; Lopez BL; Christopher TA; Wang C; Lee JC; Feuerstein GZ; Yue TL Inhibition of p38 mitogen-activated protein kinase decreases cardiomyocyte apoptosis and improves cardiac function after myocardial ischemia and reperfusion. Circulation 1999, 99 (13), 1685–1691. DOI: 10.1161/01.cir.99.13.1685 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (38).Zhang W; Elimban V; Nijjar MS; Gupta SK; Dhalla NS Role of mitogen-activated protein kinase in cardiac hypertrophy and heart failure. Exp Clin Cardiol 2003, 8 (4), 173–183. [PMC free article] [PubMed] [Google Scholar]
- (39).Braz JC; Bueno OF; De Windt LJ; Molkentin JD PKC alpha regulates the hypertrophic growth of cardiomyocytes through extracellular signal-regulated kinase1/2 (ERK1/2). J Cell Biol 2002, 156 (5), 905–919. DOI: 10.1083/jcb.200108062 From NLM Medline. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (40).Schreckenberg R; Taimor G; Piper HM; Schluter KD Inhibition of Ca2+-dependent PKC isoforms unmasks ERK-dependent hypertrophic growth evoked by phenylephrine in adult ventricular cardiomyocytes. Cardiovasc Res 2004, 63 (3), 553–560. DOI: 10.1016/j.cardiores.2004.04.032 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (41).Allo SN; Carl LL; Morgan HE Acceleration of growth of cultured cardiomyocytes and translocation of protein kinase C. Am J Physiol 1992, 263 (2 Pt 1), C319–325. DOI: 10.1152/ajpcell.1992.263.2.C319 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (42).Dunnmon PM; Iwaki K; Henderson SA; Sen A; Chien KR Phorbol esters induce immediate-early genes and activate cardiac gene transcription in neonatal rat myocardial cells. J Mol Cell Cardiol 1990, 22 (8), 901–910. DOI: 10.1016/0022-2828(90)90121-h From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (43).Solan JL; Lampe PD Key connexin 43 phosphorylation events regulate the gap junction life cycle. J Membr Biol 2007, 217 (1–3), 35–41. DOI: 10.1007/s00232-007-9035-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (44).Kolettis TM Endothelin-1 during myocardial ischaemia: a double-edged sword? Hypertens Res 2011, 34 (2), 170–172. DOI: 10.1038/hr.2010.215 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (45).Archer CR; Robinson EL; Drawnel FM; Roderick HL Endothelin-1 promotes hypertrophic remodelling of cardiac myocytes by activating sustained signalling and transcription downstream of endothelin type A receptors. Cell Signal 2017, 36, 240–254. DOI: 10.1016/j.cellsig.2017.04.010 From NLM Medline. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (46).Peng C; Luo X; Li S; Sun H Phenylephrine-induced cardiac hypertrophy is attenuated by a histone acetylase inhibitor anacardic acid in mice. Mol Biosyst 2017, 13 (4), 714–724. DOI: 10.1039/c6mb00692b From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (47).Kong Y; Shi W; Zhang D; Jiang X; Kuss M; Liu B; Li Y; Duan B Injectable, antioxidative, and neurotrophic factor-deliverable hydrogel for peripheral nerve regeneration and neuropathic pain relief. Applied Materials Today 2021, 24, 101090. DOI: 10.1016/j.apmt.2021.101090. [DOI] [Google Scholar]
- (48).Zhang W; Gilstrap K; Wu L; K. C RB; Moss MA; Wang Q; Lu X; He X Synthesis and Characterization of Thermally Responsive Pluronic F127−Chitosan Nanocapsules for Controlled Release and Intracellular Delivery of Small Molecules. ACS Nano 2010, 4 (11), 6747–6759. DOI: 10.1021/nn101617n. [DOI] [PubMed] [Google Scholar]
- (49).Kong Y; Shi W; Zheng L; Zhang D; Jiang X; Liu B; Xue W; Kuss M; Li Y; Sorgen PL; et al. In situ delivery of a curcumin-loaded dynamic hydrogel for the treatment of chronic peripheral neuropathy. Journal of Controlled Release 2023, 357, 319–332. DOI: 10.1016/j.jconrel.2023.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (50).Shi W; Fang F; Kong Y; Greer SE; Kuss M; Liu B; Xue W; Jiang X; Lovell P; Mohs AM; et al. Dynamic hyaluronic acid hydrogel with covalent linked gelatin as an anti-oxidative bioink for cartilage tissue engineering. Biofabrication 2022, 14 (1), 014107. DOI: 10.1088/1758-5090/ac42de. [DOI] [PubMed] [Google Scholar]
- (51).Wang L; Qing L; Liu H; Liu N; Qiao J; Cui C; He T; Zhao R; Liu F; Yan F; et al. Mesenchymal stromal cells ameliorate oxidative stress-induced islet endothelium apoptosis and functional impairment via Wnt4-beta-catenin signaling. Stem Cell Res Ther 2017, 8 (1), 188. DOI: 10.1186/s13287-017-0640-0 From NLM Medline. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (52).Solan JL; Marquez-Rosado L; Sorgen PL; Thornton PJ; Gafken PR; Lampe PD Phosphorylation at S365 is a gatekeeper event that changes the structure of Cx43 and prevents down-regulation by PKC. J Cell Biol 2007, 179 (6), 1301–1309. DOI: 10.1083/jcb.200707060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (53).Dhaun N; Webb DJ Endothelins in cardiovascular biology and therapeutics. Nat Rev Cardiol 2019, 16 (8), 491–502. DOI: 10.1038/s41569-019-0176-3 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (54).Miyauchi T; Sakai S Endothelin and the heart in health and diseases. Peptides 2019, 111, 77–88. DOI: 10.1016/j.peptides.2018.10.002 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (55).Gan XT; Rajapurohitam V; Haist JV; Chidiac P; Cook MA; Karmazyn M Inhibition of phenylephrine-induced cardiomyocyte hypertrophy by activation of multiple adenosine receptor subtypes. J Pharmacol Exp Ther 2005, 312 (1), 27–34. DOI: 10.1124/jpet.104.073122 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (56).Kemp TJ; Aggeli IK; Sugden PH; Clerk A Phenylephrine and endothelin-1 upregulate connective tissue growth factor in neonatal rat cardiac myocytes. J Mol Cell Cardiol 2004, 37 (2), 603–606. DOI: 10.1016/j.yjmcc.2004.04.022 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (57).Li P; Nijhawan D; Budihardjo I; Srinivasula SM; Ahmad M; Alnemri ES; Wang X Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell 1997, 91 (4), 479–489. DOI: 10.1016/s0092-8674(00)80434-1 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (58).Kim SJ Global Awareness of Myocardial Infarction Symptoms in General Population. Korean Circ J 2021, 51 (12), 997–1000. DOI: 10.4070/kcj.2021.0320 From NLM PubMed-not-MEDLINE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (59).Dhein S; Salameh A Remodeling of Cardiac Gap Junctional Cell-Cell Coupling. Cells 2021, 10 (9). DOI: 10.3390/cells10092422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (60).Ruschitzka F; Abraham WT; Singh JP; Bax JJ; Borer JS; Brugada J; Dickstein K; Ford I; Gorcsan J 3rd; Gras D; et al. Cardiac-resynchronization therapy in heart failure with a narrow QRS complex. N Engl J Med 2013, 369 (15), 1395–1405. DOI: 10.1056/NEJMoa1306687 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (61).Palandri C; Santini L; Argiro A; Margara F; Doste R; Bueno-Orovio A; Olivotto I; Coppini R Pharmacological Management of Hypertrophic Cardiomyopathy: From Bench to Bedside. Drugs 2022, 82 (8), 889–912. DOI: 10.1007/s40265-022-01728-w From NLM Medline. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (62).Rurik JG; Aghajanian H; Epstein JA Immune Cells and Immunotherapy for Cardiac Injury and Repair. Circ Res 2021, 128 (11), 1766–1779. DOI: 10.1161/CIRCRESAHA.121.318005 From NLM Medline. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (63).Silva AC; Pereira C; Fonseca A; Pinto-do OP; Nascimento DS Bearing My Heart: The Role of Extracellular Matrix on Cardiac Development, Homeostasis, and Injury Response. Front Cell Dev Biol 2020, 8, 621644. DOI: 10.3389/fcell.2020.621644 From NLM PubMed-not-MEDLINE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (64).Johnson T; Zhao L; Manuel G; Taylor H; Liu D Approaches to therapeutic angiogenesis for ischemic heart disease. J Mol Med (Berl) 2019, 97 (2), 141–151. DOI: 10.1007/s00109-018-1729-3 From NLM Medline. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (65).Hashimoto H; Olson EN; Bassel-Duby R Therapeutic approaches for cardiac regeneration and repair. Nat Rev Cardiol 2018, 15 (10), 585–600. DOI: 10.1038/s41569-018-0036-6 From NLM Medline. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (66).Marsh SR; Williams ZJ; Pridham KJ; Gourdie RG Peptidic Connexin43 Therapeutics in Cardiac Reparative Medicine. J Cardiovasc Dev Dis 2021, 8 (5). DOI: 10.3390/jcdd8050052 From NLM PubMed-not-MEDLINE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (67).Smith JH; Green CR; Peters NS; Rothery S; Severs NJ Altered patterns of gap junction distribution in ischemic heart disease. An immunohistochemical study of human myocardium using laser scanning confocal microscopy. Am J Pathol 1991, 139 (4), 801–821. [PMC free article] [PubMed] [Google Scholar]
- (68).Pun R; Kim MH; North BJ Role of Connexin 43 phosphorylation on Serine-368 by PKC in cardiac function and disease. Front Cardiovasc Med 2022, 9, 1080131. DOI: 10.3389/fcvm.2022.1080131 From NLM PubMed-not-MEDLINE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (69).Schulz R; Gorge PM; Gorbe A; Ferdinandy P; Lampe PD; Leybaert L Connexin 43 is an emerging therapeutic target in ischemia/reperfusion injury, cardioprotection and neuroprotection. Pharmacol Ther 2015, 153, 90–106. DOI: 10.1016/j.pharmthera.2015.06.005 From NLM Medline. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (70).Jiang J; Hoagland D; Palatinus JA; He H; Iyyathurai J; Jourdan LJ; Bultynck G; Wang Z; Zhang Z; Schey K; et al. Interaction of alpha Carboxyl Terminus 1 Peptide With the Connexin 43 Carboxyl Terminus Preserves Left Ventricular Function After Ischemia-Reperfusion Injury. J Am Heart Assoc 2019, 8 (16), e012385. DOI: 10.1161/JAHA.119.012385 From NLM Medline. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (71).O’Quinn MP; Palatinus JA; Harris BS; Hewett KW; Gourdie RG A peptide mimetic of the connexin43 carboxyl terminus reduces gap junction remodeling and induced arrhythmia following ventricular injury. Circ Res 2011, 108 (6), 704–715. DOI: 10.1161/CIRCRESAHA.110.235747 From NLM Medline. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (72).Mukoyama M; Nakao K; Obata K; Jougasaki M; Yoshimura M; Morita E; Hosoda K; Suga S; Ogawa Y; Yasue H; et al. Augmented secretion of brain natriuretic peptide in acute myocardial infarction. Biochem Biophys Res Commun 1991, 180 (1), 431–436. DOI: 10.1016/s0006-291x(05)81311-7 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (73).Lainchbury JG; Espiner EA; Frampton CM; Richards AM; Yandle TG; Nicholls MG Cardiac natriuretic peptides as predictors of mortality. J Intern Med 1997, 241 (4), 257–259. DOI: 10.1046/j.1365-2796.1997.295134000.x From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (74).Foy SG; Crozier IG; Richards AM; Nicholls MG; Turner JG; Frampton CM; Ikram H Neurohormonal changes after acute myocardial infarction. Relationships with haemodynamic indices and effects of ACE inhibition. Eur Heart J 1995, 16 (6), 770–778. DOI: 10.1093/oxfordjournals.eurheartj.a060995 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (75).Akazawa H; Komuro I Roles of cardiac transcription factors in cardiac hypertrophy. Circ Res 2003, 92 (10), 1079–1088. DOI: 10.1161/01.RES.0000072977.86706.23 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (76).Kessler-Icekson G; Barhum Y; Schaper J; Schaper W; Kaganovsky E; Brand T ANP expression in the hypertensive heart. Exp Clin Cardiol 2002, 7 (2–3), 80–84. [PMC free article] [PubMed] [Google Scholar]
- (77).Ginnan R; Singer HA CaM kinase II-dependent activation of tyrosine kinases and ERK1/2 in vascular smooth muscle. Am J Physiol Cell Physiol 2002, 282 (4), C754–761. DOI: 10.1152/ajpcell.00335.2001 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (78).Sun Y; Chen Y; Zhan L; Zhang L; Hu J; Gao Z The role of non-receptor protein tyrosine kinases in the excitotoxicity induced by the overactivation of NMDA receptors. Rev Neurosci 2016, 27 (3), 283–289. DOI: 10.1515/revneuro-2015-0037 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (79).Khera L; Vinik Y; Maina F; Lev S The AXL-PYK2-PKCalpha axis as a nexus of stemness circuits in TNBC. Life Sci Alliance 2021, 4 (6). DOI: 10.26508/lsa.202000985 From NLM Medline. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (80).Blaukat A; Ivankovic-Dikic I; Gronroos E; Dolfi F; Tokiwa G; Vuori K; Dikic I Adaptor proteins Grb2 and Crk couple Pyk2 with activation of specific mitogen-activated protein kinase cascades. J Biol Chem 1999, 274 (21), 14893–14901. DOI: 10.1074/jbc.274.21.14893 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (81).Miao LN; Pan D; Shi J; Du JP; Chen PF; Gao J; Yu Y; Shi DZ; Guo M Role and Mechanism of PKC-delta for Cardiovascular Disease: Current Status and Perspective. Front Cardiovasc Med 2022, 9, 816369. DOI: 10.3389/fcvm.2022.816369 From NLM PubMed-not-MEDLINE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (82).Gallo S; Vitacolonna A; Bonzano A; Comoglio P; Crepaldi T ERK: A Key Player in the Pathophysiology of Cardiac Hypertrophy. Int J Mol Sci 2019, 20 (9). DOI: 10.3390/ijms20092164 From NLM Medline. [DOI] [PMC free article] [PubMed] [Google Scholar]
- (83).Yoshizumi M; Abe J; Haendeler J; Huang Q; Berk BC Src and Cas mediate JNK activation but not ERK1/2 and p38 kinases by reactive oxygen species. J Biol Chem 2000, 275 (16), 11706–11712. DOI: 10.1074/jbc.275.16.11706 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (84).Zhang C; Hsieh MH; Wu SY; Li SH; Wu J; Liu SM; Wei HJ; Weisel RD; Sung HW; Li RK A self-doping conductive polymer hydrogel that can restore electrical impulse propagation at myocardial infarct to prevent cardiac arrhythmia and preserve ventricular function. Biomaterials 2020, 231, 119672. DOI: 10.1016/j.biomaterials.2019.119672 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (85).Ruvinov E; Cohen S Alginate biomaterial for the treatment of myocardial infarction: Progress, translational strategies, and clinical outlook: From ocean algae to patient bedside. Adv Drug Deliv Rev 2016, 96, 54–76. DOI: 10.1016/j.addr.2015.04.021 From NLM Medline. [DOI] [PubMed] [Google Scholar]
- (86).Sack KL; Aliotta E; Choy JS; Ennis DB; Davies NH; Franz T; Kassab GS; Guccione JM Intra-myocardial alginate hydrogel injection acts as a left ventricular mid-wall constraint in swine. Acta Biomater 2020, 111, 170–180. DOI: 10.1016/j.actbio.2020.04.033 From NLM Medline. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.