1. Introduction
Cancer remains a major global health challenge, with approximately 19.3 million new cases and 10 million deaths reported in 2020, and this number is expected to rise to 28.4 million new cases annually by 2040 [
1]. Nanohydrogels represent a novel class of nanomaterials characterized by their high water content and biocompatibility, emerging as effective drug delivery systems in cancer therapy [
2,
3]. Their hydrophilic polymer networks facilitate the encapsulation of various therapeutic agents, enhancing the solubility of hydrophobic drugs and minimizing systemic toxicity [
4,
5]. Specifically, DNA nanohydrogels leverage the unique structural and functional properties of DNA to provide superior programmability and biocompatibility [
6,
7] in order to tailor drug release. The programmability renders DNA nanohydrogels particularly suitable for the delivery of genetic therapeutics [
8,
9], thereby achieving dual effects [
10] to expand the scopes of conventional chemo-drugs.
However, precise spatial and temporal manipulation of drug release remains a significant challenge in the development of DNA nanohydrogels. Overcoming these challenges is essential for the advancement of smart drug delivery systems that can optimize therapeutic outcomes. Currently, a variety of stimuli-responsive nanomaterials, including polymeric nanoparticles [
11], liposomes [
12], inorganic nanocarriers [
13], and micelles [
14], have been explored for drug delivery applications. These materials can react to specific stimuli such as pH, temperature, light, or biochemical signals, enabling targeted drug delivery and on-demand release, which can enhance therapeutic efficacy and reduce off-target effects [
15]
. However, issues related to potential toxicity and intricate fabrication processes may restrict their broader applications. Recent advances in smart DNA functionalization strategies have demonstrated that intelligent design of DNA-based systems can significantly enhance drug delivery efficiency by enabling precise control over drug release kinetics and targeting specificity [
16,
17].
Consequently, there is an urgent need to develop DNA nanohydrogels that incorporate these responsive features. DNA nanohydrogels exhibit remarkable programmability, allowing for the design of systems responsive to specific biological stimuli, such as pH, mRNA, or specific biomolecules within tumor microenvironment [
18,
19]. This responsiveness would pave the way for integrating chemotherapeutics with gene therapies [
20,
21], aligning a timely release of therapeutic agents with targeted genetic interventions. In this context, we have developed a pH/mRNA stimuli-responsive dendritic-DNA-based nanohydrogel (
DOX@pH-DNG-ASO) for the co-delivery of doxorubicin (DOX) and antisense oligonucleotides (ASOs) to achieve combined therapeutic effects in vitro (
Scheme 1). This multifunctional DNA nanohydrogel demonstrates significant anticancer activity through a combination approach of chemo–gene treatments, which involves silencing the tumor-associated gene HMGN5 [
22,
23].
2. Materials and Methods
2.1. Chemicals and Materials
All chemicals and solvents utilized were of analytical grade. Millipore water was employed to prepare all aqueous solutions. Linear DNA and RNA strands were procured from Sangon Biotech Co., Ltd. (Shanghai, China). Modified DNA strands and all RNA strands underwent purification via high-performance liquid chromatography (HPLC). All the sequences are shown in
Table S1. DEPC-treated water was used to dissolve RNA-related materials. Reagents and phosphoramidites used in solid-phase DNA syntheses were sourced from Beijing Higgrene-tech Automation Ltd. (Beijing, China). Unless otherwise specified, all chemicals were obtained from Sigma-Aldrich (St. Louis, MO, USA).
2.2. Oligonucleotide Synthesis
Dendritic DNA was synthesized using a solid-phase DNA synthesizer (K&A H-8, Schaafheim, Germany) with 2000 Å CPG, following our established protocol [
24] and characterized via mass spectrometry (
Figures S1 and S2).
2.3. Assembly of pH-DNG
The assembly of pH-DNG was conducted using a method similar to that of non-pH-DNG [
24]. A schematic representation of the assembly process is provided in
Figure S1, illustrating the key chemical routes and steps involved in the formation of pH-DNG. Dendritic DNA (100 nM) and linker (100 nM) were combined in 1× TAE/Mg
2+ buffer (containing 40 mM Tris, 20 mM acetic acid, 2 mM EDTA, and 12.5 mM Mg
2+, with the pH adjusted to 7.4). The mixture was heated to 95 °C and then gradually cooled to room temperature. Subsequently, the nanogels were dialyzed (using a 25K MWCO membrane from Yuanye Bio-Technology Co., Ltd., Shanghai, China) to eliminate excess dendritic DNA and linker oligonucleotides. The purified nanogels were then concentrated through lyophilization. The resulting pH-DNG powder was then diluted in 1× TAE/Mg
2+ buffer (pH 7.4) to prepare a stock solution (Abs260 = 0.5). This stock solution was used for subsequent experiments unless otherwise indicated.
2.4. Particle Size and Zeta Potential Measurements
The hydrodynamic size and zeta potential of the samples were determined using a Zetasizer Advance Pro (ZX XPLORER): Version 2.3.1.4 (Malvern, UK). Each sample was measured three times to ensure accuracy.
2.5. Atomic Force Microscopic (AFM)
AFM images were obtained using an atomic force microscope (MFP-3D Origin+, Oxford, UK) in tapping mode at room temperature in air. The AFM measurements were performed using AC-240TSA-R3 tips (Oxford Instruments, Santa Barbara, CA, USA) with a nominal spring constant of 2 N/m and a resonance frequency of 70 kHz. Prior to data acquisition, the probes were calibrated to ensure accuracy and reproducibility. The deflection sensitivity was determined by force–distance curves on a clean silicon surface.
2.6. DOX Loading of pH-DNG
According to our previous work [
24], doxorubicin (DOX) solution in 12.5 mM MgCl
2, 1× TAE buffer (pH 7.4) was first shaken continuously with
pH-DNG (0.5 OD) at room temperature for 24 h in the darkness. After the loading process, excess free DOX was removed by dialysis bag (MW = 1000 Da, Yuanye Bio-Technology Co., Ltd., Shanghai) for 24 h in the darkness. Then the concentration of DOX in the dialysate ([DOX]_out) was quantified by a UV–vis spectrometer (Shimadzu UV-2600, Kyoto, Japan), according to the extinction coefficient of DOX at 480 nm (ε DOX = 11,500). The loading number of DOX per base pair (
RDOX) was calculated using the following formula:
where [bp] is the base pair concentration of the
pH-DNG solution inside the dialysis bag, determined by the original OD of
pH-DNG. The loading experiments were averaged over three repeats.
2.7. Doxorubicin Release
A total of 100 mL of 1× PBS buffer (pH 7.4 or 5.5) was used as drug release media. A total of 500 μL of DOX@pH-DNG was encapsulated within a dialysis bag (MW = 1000 Da) and stirred in 1× PBS buffer at pH 7.4 or 5.5. At the selected time intervals, 3 μL of DOX@pH-DNG was taken out to quantify the DOX concentration inside the dialysis membrane by UV–vis absorption spectroscopy (NanoPhotometer N60, IMPLEN, München, Germany) at 480 nm.
2.8. Assembly of pH-DNG-ASO
Initially, dendritic oligonucleotides (100 nM), linker strands (100 nM), shielding strands (100 nM), and ASO strands (100 nM) were mixed in 1 mL of 12.5 mM MgCl2, 1× TAE buffer (pH = 7.4). The mixture was heated to 95 °C and slowly cooled down to room temperature overnight. The resulting nanohydrogels were dialyzed (MW = 25,000 Da, Yuanye Bio-Technology Co., Ltd., Shanghai) to remove the excess oligonucleotides. Then, purified nanohydrogels were concentrated by lyophilization. A total of 100 μL DEPC water was added to make stock solution. Then, the stock solution was diluted to make pH-DNG-ASO (0.5 OD).
2.9. ASO Release from pH-DNG-ASO
The pH-DNG-ASO samples were incubated with target mRNA or scrambled RNA in 1× TAE buffer (pH 7.0) for 4 h at 37 °C. Then, the samples were characterized by 10% native polyacrylamide gel electrophoresis (PAGE) (running buffer: 1× TBE; running voltage: 350 V).
2.10. Doxorubicin Loading of pH-DNG-ASO
The procedure for doxorubicin loading onto pH-DNG-ASO is identical to that used for pH-DNG.
2.11. Cell Culture
The human breast cancer cell line MDA-MB-231 cells (catalog number TCHu227) were routinely cultured in high-glucose DMEM supplemented with 10% (v/v) fetal bovine serum (FBS) and 1% (w/v) penicillin–streptomycin under 5% CO2 at 37 °C. The human mammary epithelial cell line MCF-10A cells (catalog number SCSP-575) were routinely cultured in MCF-10A-specific medium under 5% CO2 at 37 °C. The human lung adenocarcinoma cell line A549 (catalog number: TCHu150), a model for non-small-cell lung cancer (NSCLC), was routinely cultured in RPMI-1640 supplemented with 10% (v/v) fetal bovine serum (FBS) and 1% (w/v) penicillin–streptomycin under 5% CO2 at 37 °C. The cancer cells were obtained from the National Collection of Authenticated Cell Cultures (NCACC).
2.12. Confocal Fluorescence Microscopy Imaging
MDA-MB-231 cells (2 × 105 per well) were seeded in a 20 mm u-dish containing DMEM cell medium. pH-TAMRA-labeled pH-DNG, DOX@pH-DNG, Cy5-labeled pH-DNG-ASO, and DOX@pH-DNG-ASO were added to the cell medium and incubated for 4 h, respectively. Afterwards, the cell medium was removed and the cells were washed with PBS for three times. Next, the cells were fixed with 4% paraformaldehyde for 15 min and stained with DAPI according to commercial protocol. The cell medium was replaced by PBS before the confocal images were taken. All images were collected on a confocal microscope (Zeiss LSM 880) with a 40 × oil immersion objective. For DOX and TAMRA, a 488 nm laser was used for excitation, with an emission range of 580 ± 20 nm. For Cy5.5, a 633 nm laser was used for excitation, with an emission range of 668 ± 20 nm.
2.13. Flow Cytometry
MDA-MB-231 cells (8 × 105 per well) were plated in 35 mm cell culture dishes and incubated overnight. The cells were then incubated with free DOX, DOX@pH-DNG, pH-DNG-ASO, and naked ASOs for 4 h, respectively. Following incubation, the cells were finally suspended in 400 μL PBS for flow cytometry analysis using a NovoCyte instrument (ACEA, San Diego, CA, USA). The untreated cells were used as negative control.
2.14. Lysosomal Colocalization of pH-DNG-ASO
MDA-MB-231 cells were seeded into confocal dishes (2 × 105 cells per well) and incubated with Cy5-labeled pH-DNG-ASO for different durations (4 and 12 h). The cells were stained with LysoTracker Green for 2 h and Hoechst 33342 for 15 min. Then, the cells were washed three times with PBS and imaged by CLSM (LSM 880, Zeiss, Oberkochen, Germany). For LysoTracker Green, the excitation wavelength was 488 nm, and the emission range was 500–550 nm.
2.15. Cell Viability Assay
The MDA-MB-231 cells were seeded at a density of 1.0 × 104 cells per well into the 96-well plates and cultured overnight. Then, the cells were, respectively, incubated with pH-DNG, free DOX, DOX@pH-DNG, DOX@non-pH-DNG, pH-DNG-ASO, and DOX@pH-DNG-ASO at different concentrations for 48 h. After incubation, CCK-8 solution was added to each group for continued culturing for 1 h. The absorption at 450 nm was measured using a microplate reader (SPARK, Tecan, Mount Waverley, Australia).
The A549 cells were seeded at a density of 1.0 × 104 cells per well into the 96-well plates and cultured overnight. Then, the cells were, respectively, incubated with free DOX, DOX@non-pH-DNG, and DOX@pH-DNG at different DOX concentrations for 48 h. After incubation, CCK-8 solution was added to each group for continued culture for 1 h. The absorption at 450 nm was measured using a microplate reader (SPARK, Tecan, Australia).
2.16. Scratch Healing Assay
MDA-MB-231 cells were seeded into 6-well plates (5 × 105 cells per well) and cultured at 37 °C overnight. A scratch was then made through the cell layer using a pipette tip along a straight line guided by a ruler. After scratching, the non-adherent cells were removed by washing with PBS three times. The cells were incubated with PBS, pH-DNG (0.5 OD), and pH-DNG-ASO (60 nM) for 0 and 24 h, respectively. Then, the change in the gap area was observed under a microscope (Leica, Wetzlar, Germany).
2.17. Quantitative Reverse Transcription Polymerase Chain Reaction (qRT-PCR)
The MDA-MB-231 cells were seeded into the six-well plates (2 × 10
5 cells per well) and incubated with PBS and
pH-DNG-ASO for 24 and 48 h, respectively. Then, the cells were washed three times with PBS. Total mRNA was extracted with Trizol agent according to the manufacturer’s instructions. Then the equal amounts of RNA were reverse-transcribed to complementary DNA (cDNA) by using a commercial kit. After that, the PCR process was carried out by mixing primers, cDNA, dNTP, and DNA polymerase. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was amplified as an internal control. The primer sequences are shown in
Table S1.
2.18. Western Blot
Cells were lysed using RIPA buffer supplemented with 1× protease inhibitor cocktail and 1 mM PMSF. The lysates were centrifuged at 12,000× g for 15 min at 4 °C to remove cellular debris. The supernatant was collected, and protein concentrations were determined using a BCA protein assay kit (Beyotime, Shanghai, China, Cat No. P0010S) according to the manufacturer’s instructions. Equal amounts of protein (20 µg per sample) were heated at 100 °C for 5 min to denature the proteins. Samples were then loaded onto 4–12% Bis-Tris precast gels alongside a pre-stained protein marker (6.5–270 kDa). Electrophoresis was performed using MOPS running buffer at 120 V for approximately 90 min or until the dye front reached the bottom of the gel. Proteins were then transferred from the gel to a PVDF membrane (Millipore, St. Louis, MO, USA, Cat No. IPVH00010) using a wet transfer system (Bio-Rad) at 100 V for 90 min in transfer buffer containing 25 mM Tris, 192 mM glycine, and 20% methanol. The membrane was blocked with 5% non-fat dry milk (Bio-Rad, Singapore, Cat No. 1706404) in TBST (20 mM Tris, 150 mM NaCl, 0.1% Tween-20, pH 7.6) for 1 h at room temperature to prevent non-specific binding. After blocking, the membrane was incubated overnight at 4 °C with the primary antibody against HMGN5 (NSBP1 Polyclonal Antibody, Proteintech, Singapore, Cat No. 23955-1-AP, 1:1000 dilution in 5% BSA/TBST). Following primary antibody incubation, the membrane was washed three times with TBST (10 min each) and incubated with a horseradish peroxidase (HRP)-conjugated secondary antibody (anti-rabbit IgG, Cell Signaling Technology, Cat No. 7074S, 1:5000 dilution in 5% non-fat dry milk/TBST) for 1 h at room temperature. After washing the membrane three times with TBST (10 min each), protein bands were visualized using an enhanced chemiluminescence (ECL) detection kit (Thermo Fisher Scientific, Singapore, Cat No. 32106) according to the manufacturer’s instructions. Images were captured using Odyssey® Fc (Licor, Bourne, MA, USA). GAPDH (Cell Signaling Technology, Singapore, Cat No. 5174S, 1:5000 dilution) was used as a loading control.
2.19. Statistical Analysis
Statistical analysis results are presented as means ± standard deviation. Data were analyzed using one-way analysis of variance (ANOVA) in GraphPad Prism 8.0 for comparisons involving more than two groups. Statistical significance was recognized at p < 0.05. Asterisks represent significant differences (* p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001).
2.20. Software Tools
In this study, multiple software tools were employed for data processing: AFM image data were processed using ImageJ software, which enabled operations such as length analysis to determine the size of nanohydrogels. DLS data were processed using Zetasizer software to analyze the particle size distribution and zeta potential of nanohydrogels. Confocal fluorescence microscopy images were acquired and initially processed using ZEN software (Version 3.4.91.00000, integrated with Zeiss LSM 880), with further colocalization analysis performed using ImageJ software. Data plotting was performed using OriginPro (Version 2021 (9.8.0.200)), while statistical analyses were conducted using GraphPad Prism.
4. Discussion
Designing intelligent nanodelivery systems responsive to the tumor microenvironment (TME) is crucial for enhancing the precision and efficacy of cancer therapy. This study introduces a pH/ASO dual-responsive DNA nanohydrogel (DOX@pH-DNG-ASO) that integrates chemotherapeutic and gene therapy modules, demonstrating combined anticancer potential. The efficient drug loading and responsive mechanisms highlight the versatility of DNA nanomaterials in precision medicine.
pH-DNG forms spherical nanoparticles (~335 nm) with a near-neutral surface charge (−7.3 mV), optimizing tumor targeting via the enhanced permeability and retention (EPR) effect and minimizing nonspecific interactions. The modular design of dendritic DNA allows for ASO incorporation without disrupting the hydrogel matrix, enabling versatile cargo integration. The nanohydrogel rapidly disintegrates at pH 5.5 due to i-motif collapse, ensuring TME-specific drug release. DOX exhibits minimal leakage at physiological pH but rapid release in the acidic TME, maximizing accumulation at the target site while reducing off-target toxicity. This pH-dependent behavior leverages DNA hybridization for superior programmability compared to traditional carriers. ASO release is mediated by toehold-driven strand displacement, ensuring specificity for target
HMGN5 mRNA. This mechanism minimizes off-target effects, a common limitation of conventional antisense therapies. Efficient cytoplasmic delivery and lysosomal escape further enhance ASO functionality, overcoming biological barriers to nucleic acid delivery. The combined
DOX@pH-DNG-ASO system reduces the IC
50 by 40-fold compared to free DOX in MDA-MB-231 cells, attributed to TME-specific DOX accumulation and ASO-mediated HMGN5 silencing. Additionally,
pH-DNG also enhanced DOX cytotoxicity in A549 cells, particularly at low drug concentrations. These results demonstrate the effectiveness of its pH-responsive release mechanism across different cancer types. In MDA-MB-231 cells, scratch assaying and qRT-PCR data show that ~70%
HMGN5 mRNA downregulation inhibits cancer cell migration, consistent with its role in breast cancer progression. Moreover, Western blot analysis verified that pH—DNG—ASO inhibited HMGN5 at the translational level by reducing its protein levels. In this combined treatment, gene therapy may enhance the efficacy of chemotherapy through the following mechanisms: For chemosensitization, knocking down
HMGN5 could impair DNA repair pathways such as homologous recombination, thereby potentiating DOX-induced DNA damage [
27]. For drug resistance mitigation,
HMGN5 has been linked to multidrug resistance (MDR) through the regulation of ABC transporters, and its silencing may reverse chemoresistance [
28].
Overall, this “chemo-gene” strategy enhances therapeutic efficacy while reducing required drug doses, aligning with the trends in combinatorial nanomedicine. However, the in vivo stability, biodistribution, and immune compatibility of pH-DNG require further validation. Future studies could explore broader mRNA targets, optimize ASO sequences, extend gene therapy applications to NSCLC-associated targets in A549 cells, test efficacy in additional cancer models, and evaluate clinical potential to advance pH-DNG as a universal combination platform.