We are exploring the versatile potential of electrospun nanofibers in various biomedical applications, particularly in wound healing, tissue engineering, and bone regeneration. Nanofibers, due to their high surface area, porosity, and ease of functionalization, are ideal candidates for creating scaffolds and drug delivery systems that mimic natural extracellular matrices, providing excellent support for cell growth and tissue regeneration.
The development of effective cancer therapies requires innovative approaches to deliver drugs precisely to tumor sites while minimizing harm to surrounding healthy tissues. In our lab, we focus on advancing targeted drug delivery systems that leverage nanotechnology to enhance the efficacy and specificity of cancer treatments, particularly for metastatic breast cancer (MBC). Our research is centered on the use of multifunctional liposomes and pH-sensitive formulations designed to overcome the limitations of traditional chemotherapy, such as systemic toxicity, non-specific targeting, and poor drug bioavailability.
Breast cancer (BC) is the second most prevalent form of cancer worldwide. Minimal residual disease (MRD), the small population of tumor cells left near the tumor after surgical ablation, leads to tumor recurrence. Overexpression of efflux proteins makes the MRD resistant to taxane therapy. Paclitaxel (PTX) is the first-line chemotherapeutic agent in BC. However, prolonged administration results in the emergence of multidrug resistance due to the expression of P-glycoprotein and metastasis. Simvastatin (SIM), a cholesterol synthesis inhibitor, has shown tremendous anticancer potential in a plethora of cancers along with pronounced anti-invasive activity and P-glycoprotein downregulation. In order to prevent untoward adverse effects, nanofiber implants loaded with both drugs were fabricated with sustained release for up to 7 days. Enhanced apoptosis was observed for drugs post-incorporation into the nanofibers with prominent anti-invasive activity. The incorporation of simvastatin resulted in P-glycoprotein efflux reversal of paclitaxel as evidenced by the rhodamine 123 assay and PTX sensitivity. The transwell invasion assay reduced the number of invading cells post-fiber treatment. Thus, we conclude that PTX- and SIM-loaded nanofibers could be a viable therapeutic alternative to prevent post-surgical tumor recurrence and cancer cell invasion.
Oral carcinoma (OC) is the leading cause of mouth deformation and mortality in the world. It is the 6th most frequent cancer globally. OC is aggressive cancer-forming tumors that invade nearby tissues, leading to locoregional spread. Surgical ablation of large tumors leads to removal of a large chunk of the mouth leading to loss of function and aesthetics. Hence, the use of neoadjuvant and adjuvant chemotherapy is common for the shrinking of tumors and the prevention of tumor recurrence. In this research, we have developed niclosamide (NIC)-loaded nanoparticles-impregnated gelatin nanofibers (NNPF) for local action. The developed nanoparticles demonstrated superior cytotoxicity in the oral cancer cell line compared to the free drug. Cell staining studies demonstrated increased apoptosis and reactive oxygen species generation after treatment with NIC-loaded nanoparticles (NNP). Cell cycle analysis demonstrated cell cycle inhibition in the G0/G1 phase. Cytotoxicity studies in 3D tumoroids demonstrated the ability of NNP to prevent cancer cell growth in 3D arrangement. Thus, NNPF could prove to be a boon for localized delivery as implant-based systems for sustained release in various cancer therapeutic applications.
Multidrug resistance (MDR) infectious wounds are a major concern due to drug resistance, leading to increased patient morbidity. Lichenysin (LCN), a lipopeptide and biosurfactant obtained from certain strains of Bacillus licheniformis, has demonstrated an excellent antimicrobial property. The present study focuses on the fabrication and comprehensive evaluation of LCN-incorporated poly(vinyl alcohol) (PVA)/polycaprolactone (PCL)-based nanofiber scaffolds using an electrospinning technique as a potential wound healing biomaterial for the treatment of MDR infectious wounds in diabetic rats. The LCN-loaded PVA–PCL nanofiber scaffolds were characterized for their physicochemical, antimicrobial, in vitro cell line on L-929, hemocompatibility, flow cytometry, in vivo infectious wound healing, and enzyme-linked immuno sorbent assay (ELISA). Morphological analysis via scanning electron microscopy (SEM) images confirmed smooth and porous nanofibers with diameters in the range 200–300 nm. Fourier transform infrared and X-ray diffraction (XRD) results demonstrated the structural integrity, chemical compatibility, and amorphous nature of developed scaffolds. The scaffolds loaded with LCN demonstrated excellent water retention, moderate biodegradability, and sustained release of LCN for up to 72 h. Mechanical characterization demonstrated a robust tensile strength conducive to wound healing applications. Antimicrobial activity against Pseudomonas aeruginosa (P. aeruginosa) and Staphylococcus aureus (S. aureus) showed substantial antibacterial and antibiofilm activity. In vitro cell line studies showed enhanced cell adhesion, proliferation, migration, and viability, signifying the cytocompatibility of these scaffolds. In vivo studies demonstrated exceptional infectious wound healing potential in diabetic rats. These findings indicate that LCN-enriched PVA–PCL scaffolds hold significant potential as a therapeutic strategy for the treatment of MDR infectious wounds in diabetic rats through a multifaceted approach.
To address post-surgical recurrence and chemoresistance in prostate cancer, this study developed an electrospun nanofiber-based delivery system co-loaded with docetaxel (DTX) and niclosamide (NIC). Niclosamide, known for its ability to inhibit the PI3K-AKT signaling pathway, was utilized to re-sensitize DTX-resistant PC-3 cells. The sustained release profile of the DTX–NIC nanofibers demonstrated enhanced cytotoxicity, increased ROS generation, and apoptosis in resistant cancer cells. The findings suggest that this dual-drug nanofiber platform holds promise as a localized therapeutic strategy for overcoming resistance and improving clinical outcomes in prostate cancer.
This study presents the development of bilayered electrospun nanofiber mats composed of PVP and PVA, enriched with collagen and co-loaded with resveratrol (RSV) and ampicillin (AMP), aimed at accelerating burn wound repair. The nanofibers exhibited smooth morphology, excellent exudate absorption, biodegradability over two weeks, and sustained drug release. Antibacterial efficacy was confirmed against Staphylococcus aureus and Escherichia coli, along with significant biofilm inhibition. Structural analysis (XRD, FTIR) validated drug encapsulation, while hemolysis and cell viability assays demonstrated cytocompatibility and enhanced HaCaT cell proliferation. In vivo studies in a rat burn model confirmed accelerated healing, improved collagen synthesis, and reduced expression of inflammatory markers (TNF-α, IL-6). This multifunctional nanofiber system holds promise as an effective therapeutic platform for burn wound management.
Minimal residual disease at the tumor site after surgical ablation is a major cause for tumor recurrence and metastasis. The tropism of circulating tumor cells towards injured areas increases the risk of tumor recurrence. Recent studies have reported the inability of nanocarriers to penetrate the tumor site owing to short systemic half-life, high hydrostatic pressure, and inability to extravasate the tumor tissue. In this work, we developed simvastatin-loaded polycaprolactone - gelatin core-shell nanoparticles (SNP) for post-surgical management of breast cancer (BC). NP-loaded within the hyaluronic acid solution demonstrated shear thinning behavior (n = 0.3089). SNP demonstrated cytotoxic potential in the 2-D cell culture and 3-D tumoroids of MDA-MB-231 cell line in vitro. JC-1 and rhodamine-123 based assays demonstrated the ability of SNP to induce mitochondrial membrane damage. Further, cell cycle analysis revealed that the SNP prevented the progression of cell line from G0/G1 to S-phase. Simvastatin and SNP induced early apoptosis in the MDA-MB-231 cell line. Scratch, invasion, migration, and clonogenic assays demonstrated the ability of SNP to prevent cell invasion and clonal expansion. Immunocytochemistry revealed that both SIM and SNP reduced the expression of anti-apoptotic protein BCL-2 in metastatic cell line. Overall, the developed formulation demonstrates enormous potential for the post-surgery management of BC.
Human epidermal growth factor receptor-2 (HER2)-positive breast cancer metastasis remains the primary cause of mortality among women globally. Targeted therapies have revolutionized treatment efficacy, with Trastuzumab (Trast), a monoclonal antibody, targeting HER2-positive advanced breast cancer. The tumor-homing peptide iRGD enhances the intratumoral accumulation and penetration of therapeutic agents. Liposomes serve as versatile nanocarriers for both hydrophilic and hydrophobic drugs. Gefitinib (GFB) is a potential anticancer drug against HER2-positive breast cancer, while Lycorine hydrochloride (LCH) is a natural compound with anticancer and anti-inflammatory properties. This study developed TPGS-COOH-coated liposomes co-loaded with GFB and LCH, prepared by the solvent injection method, and surface-functionalized with Trast and iRGD. The dual surface-decorated liposomes (DSDLs) were characterized for their particle size (PS), polydispersity index (PDI), zeta potential (ZP), surface chemistry, surface morphology, and their crystallinity during in-vitro drug release, drug encapsulation, and in-vitro cell line studies on SK-BR-3 and MDA-MB-231 breast cancer cells. The half-maximum inhibitory concentration (IC-50) values of single decorated liposomes (SDLs), iRGD-LP, and Trast-LP, as well as DSDLs (iRGD-Trast-LP) on SK-BR-3 cells, were 6.10 ± 0.42, 4.98 ± 0.36, and 4.34 ± 0.32 μg/mL, respectively. Moreover, the IC-50 values of SDLs and DSDLs on MDA-MB-231 cells were 15.12 ± 0.68, 13.09 ± 0.59, and 11.08 ± 0.48 μg/mL, respectively. Cellular uptake studies using confocal laser scanning microscopy (CLSM) showed that iRGD and Trast functionalization significantly enhanced cellular uptake in both cell lines. The wound-healing assay demonstrated a significant reduction in SDL and DSDL-treated MDA-MB-231 cell migration compared to the control. Additionally, the blood compatibility study showed minimal hemolysis (less than 5% RBC lysis), indicating good biocompatibility and biosafety. Overall, these findings suggest that TPGS-COOH-coated, GFB and LCH co-loaded, dual-ligand (iRGD and Trast) functionalized, multifunctional liposomes could be a promising therapeutic strategy for treating HER2-positive metastatic breast cancer.
The advent of pH-sensitive liposomes (pHLips) has opened new opportunities for the improved and targeted delivery of antitumor drugs as well as gene therapeutics. Comprising fusogenic dioleylphosphatidylethanolamine (DOPE) and cholesteryl hemisuccinate (CHEMS), these nanosystems harness the acidification in the tumor microenvironment and endosomes to deliver drugs effectively. pH-responsive liposomes that are internalized through endocytosis encounter mildly acidic pH in the endosomes and thereafter fuse or destabilize the endosomal membrane, leading to subsequent cargo release into the cytoplasm. The extracellular tumor matrix also presents a slightly acidic environment that can lead to the enhanced drug release and improved targeting capabilities of the nano-delivery system. Recent studies have shown that folic acid (FA) and iRGD-coated nanocarriers, including pH-sensitive liposomes, can preferentially accumulate and deliver drugs to breast tumors that overexpress folate receptors and αvβ3 and αvβ5 integrins. This study focuses on the development and characterization of 5-Fluorouracil (5-FU)-loaded FA and iRGD surface-modified pHLips (FA-iRGD-5-FU-pHLips). The novelty of this research lies in the dual targeting mechanism utilizing FA and iRGD peptides, combined with the pH-sensitive properties of the liposomes, to enhance selective targeting and uptake by cancer cells and effective drug release in the acidic tumor environment. The prepared liposomes were small, with an average diameter of 152 ± 3.27 nm, uniform, and unilamellar, demonstrating efficient 5-FU encapsulation (93.1 ± 2.58%). Despite surface functionalization, the liposomes maintained their pH sensitivity and a neutral zeta potential, which also conferred stability and reduced aggregation. Effective pH responsiveness was demonstrated by the observation of enhanced drug release at pH 5.5 compared to physiological pH 7.4. (84.47% versus 46.41% release at pH 5.5 versus pH 7.4, respectively, in 72 h). The formulations exhibited stability for six months and were stable when subjected to simulated biological settings. Blood compatibility and cytotoxicity studies on MDA-MB-231 and SK-BR3 breast cancer cell lines revealed an enhanced cytotoxicity of the liposomal formulation that was modified with FA and iRGD compared to free 5-FU and minimal hemolysis. Collectively, these findings support the potential of FA and iRGD surface-camouflaged, pH-sensitive liposomes as a promising drug delivery strategy for breast cancer treatment.
Cardiotoxicity (CT) is a severe condition that negatively impacts heart function. β-sitosterol (BS) is a group of phytosterols and known for various pharmacological benefits, such as managing diabetes, cardiac protection, and neuroprotection. This study aims to develop niosomes (NS) containing BS, utilizing cholesterol as the lipid and Tween 80 as the stabilizer. The research focuses on designing and evaluating both conventional BS-NS and hyaluronic acid (HA) modified NS (BS-HA-NS) to enhance the specificity and efficacy of BS within cardiac tissue. The resulting niosomal formulation was spherical, with a size of about 158.51 ± 0.57 nm, an entrapment efficiency of 93.56 ± 1.48 %, and a drug loading of 8.07 ± 1.62 %. To evaluate cytotoxicity on H9c2 heart cells, the MTT assay was used. The cellular uptake of BS-NS and BS-HA-NS was confirmed by confocal microscopy on H9c2 cardiac cells. Administering BS-NS and BS-HA-NS intravenously at a dose of 10 mg/kg showed the ability to significantly decrease the levels of cardiac troponin-I (cTn-I), creatine kinase-MB (CK-MB), lactate dehydrogenase (LDH), aspartate aminotransferase (AST), and lipid peroxidation (MDA). Tissue histopathology indicated a substantial potential for repairing cardiac tissue after treatment with BS-NS and BS-HA-NS and strong cardioprotection against ISO induced myocardial tissue damages. Thus, enhancing BS's therapeutic effectiveness through niosome surface modification holds promise for mitigating cardiac damage resulting from CT.
This study presents the development of nanostructured lipid carriers (NLCs) functionalized with the pentapeptide cRGDfK for targeted delivery of gefitinib in hepatocellular carcinoma (HCC). The cRGDfK ligand specifically binds to the overexpressed αvβ3 integrin receptors on HCC cells, enhancing cellular uptake. The optimized gefitinib-loaded NLCs (cRGDfK@GF-NLC), composed of cholesterol, oleic acid, Pluronic F-68, and Phospholipon 90G, were extensively characterized using DSC and XRD. In vitro and in vivo evaluations using the HepG2 cell line revealed superior cytotoxicity, growth inhibition, and tumor-specific accumulation compared to non-functionalized NLCs and free drug. Biodistribution studies confirmed enhanced tumor targeting with minimal systemic toxicity. These findings establish cRGDfK@GF-NLC as a promising platform for targeted hepatocarcinoma therapy.
Myocardial infarction (MI) is a severe manifestation of ischemic heart disease (IHD) that impacts cardiac functions. Naringin is a bioflavonoid also known as 4-trihydroxy-flavonone-7-rhamnoglucoside, that offers numerous pharmacological benefits including antidiabetic, cardioprotective, and neuroprotective activities. Despite its benefits, the poor water solubility of naringin limits its drug delivery application. The objective of the investigation is to develop, optimize and evaluate the nanoparticles loaded with naringin (NR-LGNPs) by using lignin and Tween 80 as polymers and stabilizers, respectively. The NR-LGNPs were prepared using a modified phase separation technique while the composition as well as process variables of the NR-LGNPs were optimized using 23 factorial design of the experiment. The optimized formulation was subsequently characterized for various in vitro and in vivo parameters. The formulated optimized polymeric nanoparticles were spherical and had a size of 138.24 ± 1.36 nm with entrapment efficiency of 91.53 ± 0.84 %. The in vitro drug release was found to be 88.75 ± 1.54 % at the end of 24 h. The administration of LG-NPs at a dosage of 10 mg/kg through intravenous injection demonstrated a significant ability to mitigate the levels of creatine kinase-myoglobin binding (CK-MB), lactate dehydrogenase (LDH), aspartate aminotransferase (AST), lipid peroxidation (MDA) following in vivo animal model. The tissue histopathology study indicated the significant potential for restoration of cardiac tissue damage upon treatment with NR-LGNPs. Additionally, it was observed that the treatment with LG-NPs improved the size of the cardiac infarction, indicating its potent cardioprotective activity to combat myocardial damage induced by ISO. Thus, the enhancement of NR bioactivity through nanotechnology could potentially mitigate cardiac damage due to myocarial infarction.
Electrospinning is a versatile method for fabrication of précised nanofibrous materials for various biomedical application including tissue engineering and drug delivery. This research is aimed to fabricate the PVP/PVA nanofiber scaffold by novel electrospinning technique and to investigate the impact of process parameters (flow rate, voltage and distance) and polymer concentration/solvent combinations influence on properties of electrospun nanofibers. The in-vitro and in-vivo degradation studies were performed to evaluate the potential of electrospun PVP/PVA as a tissue engineering scaffold. The solvents used for electrospinning of PVP/PVA nanofibers were ethanol and 90% acetic acid, optimized with central composite design via Design Expert software. NF-2 and NF-35 were selected as optimised nanofiber formulation in acetic acid and ethanol, and their characterization showed diameter of 150–400 nm, tensile strength of 18.3 and 13.1 MPa, respectively. XRD data revealed the amorphous nature, and exhibited hydrophilicity (contact angles: 67.89° and 58.31° for NF-2 and NF-35). Swelling and in-vitro degradability studies displayed extended water retention as well as delayed degradation. FTIR analysis confirmed solvent-independent interactions. Additionally, hemolysis and in-vitro cytotoxicity studies revealed the non-toxic nature of fabricated scaffolds on RBCs and L929 fibroblast cells. Subcutaneous rat implantation assessed tissue response, month-long biodegradation, and biocompatibility through histological analysis of surrounding tissue. Due to its excellent biocompatibility, this porous PVP/PVA nanofiber has great potential for biomedical applications.
Psoriasis with unclear etiology and pathogenesis is one of the most prevalent chronic skin diseases. It involves excessive keratinocytes cells hyperproliferation, abnormal vascular infiltration, and differentiation as its characteristic features. Patients need therapy on a periodic basis over a long period because of psoriasis recurrence. Thus, there is a requirement for target specific drug-delivery system to treat psoriasis effectively. Therefore, we proposed the preparation of Nanostructured Lipid Carriers (NLCs) containing Tazarotene (TZT) & Calcipotriol (CPT) by the solvent-melt-emulsification process for topical delivery. Our research work involves preparation, optimization by Box Behnken Design (BBD) and characterization of TZT CPT nano-lipid-carrier formulation and further incorporation into Carbopol 934 gel base to provide better adhesion on the skin for alleviating psoriatic lesions. The prepared NLC gel was further subjected to various in vitro and in vivo evaluations. The in-vivo characterization involved MTT assay, cellular uptake study, antioxidant, and lipid profile test, cytokine profile assessment, splenomegaly measurement, weight variation, etc. In-vitro parameters such as zeta-potential (mV), particle size (nm), and percentage entrapment efficiency of TZT-CPT-NLC formulation were found to be 149.340 ± 0.340 nm, −13.14 ± 0.20 mV & 91.24 ± 7.30% respectively. SEM & TEM characterization demonstrated the spherical shape of both prepared TZT-NLC and TZT-CPT-NLC. The optimized TZT-CPT-NLC-based hydrogel showed 93.71 ± 1.11% drug release during 72 h of study, following Higuchi-model with r2 = 0.960 value as the best fit drug release kinetic model. The prepared hydrogel-based formulations exhibited optimum viscosity, pH, and spreadability for easy topical application. No skin irritation was additionally detected for the developed hydrogel formulations. The newly developed TZT-CPT-loaded-NLC gel formulation showed improved anti-psoriatic effectiveness as well as sustained release effect as compared with TZT-loaded-NLC gel, indicating that combination therapy may be a promising and viable option in topical management & treatment of psoriasis.
Current anticancer drug research includes tumor-targeted administration as a critical component because it is the best strategy to boost efficacy and decrease toxicity. Low drug concentration in cancer cells, nonspecific distribution, rapid clearance, multiple drug resistance, severe side effects, and other factors contribute to the disappointing results of traditional chemotherapy. As an innovative technique of treatments for hepatocellular carcinoma (HCC) in recent years, nanocarrier-mediated targeted drug delivery systems can overcome the aforesaid limitations via enhanced permeability and retention effect (EPR) and active targeting. Epidermal growth factor receptor (EGFR) inhibitor Gefitinib (Gefi) has dramatic effects on hepatocellular carcinoma. Herein, we developed and assessed an αvβ3 integrin receptor targeted c(RGDfK) surface modified liposomes for better targeting selectivity and therapeutic efficacy of Gefi on HCC cells. The conventional and modified Gefi loaded liposomes, i.e., denoted as Gefi-L and Gefi-c(RGDfK)-L, respectively, were prepared through the ethanol injection method and optimized via Box Behnken design (BBD). The FTIR and 1H NMR spectroscopy verified that the c(RGDfK) pentapeptides had formed an amide bond with the liposome surface. In addition, the particle size, Polydispersity index, zeta potential, encapsulation efficiency, and in-vitro Gefi release of the Gefi-L and Gefi-c(RGDfK)-L were measured and analyzed. As indicated by the MTT assay on HepG2 cells, Gefi-c(RGDfK)-L displayed considerably higher cytotoxicity than Gefi-L or Gefi alone. Throughout the incubation period, HepG2 cells took up significantly more Gefi-c(RGDfK)-L than Gefi-L. According to the in vivo biodistribution analysis, Gefi-c(RGDfK)-L accumulated more strongly at the tumor site than Gefi-L and free Gefi. Furthermore, HCC-bearing rats treated with Gefi-c(RGDfK)-L showed a substantial drop in liver marker enzymes (alanine transaminase, alkaline phosphatase, aspartate transaminase, and total bilirubin levels) compared to the disease control group. Gefi-c(RGDfK)-L suppresses tumour growth more effectively than Gefi-L and free Gefi, according to an in vivo analysis of their anticancer activities. Thus, c(RGDfK)-surface modified liposomes, i.e., Gefi-c(RGDfK)-L may serve as an efficient carrier for the targeted delivery of anticancer drugs.
Electrospun nanofibers scaffolds show promising potential in wound healing applications. This work aims to fabricate nanofibrous wound dressing as a novel approach for a topical drug delivery system. Herein, the electrospinning technique is used to design and fabricate bioabsorbable nanofibrous scaffolds of Polyvinyl alcohol/gelatin/poly (lactic-co-glycolic acid) enriched with thrombin (TMB) as hemostatic agent and vancomycin (VCM) as anti-bacterial agent for a multifunctional platform to control excessive blood loss, inhibit bacterial growth and enhance wound healing. SEM, FTIR, XRD, in vitro drug release, antimicrobial studies, biofilm, cell viability assay, and in vivo study in a rat model were used to assess nanofiber’s structural, mechanical, and biological aspects. SEM images confirms the diameter of nanofibers which falls within the range from 150 to 300 nm for all the batches. Excellent swelling index data makes it suitable to absorb wound exudates. In-vitro drug release data shows sustained release behavior of nanofiber. Nanofibers scaffolds showed biomimetic behavior and excellent biocompatibility. Moreover, scaffolds exhibited excellent antimicrobial and biofilm activity against Staphylococcus aureus. Nanofibrous scaffolds showed less bleeding time, rapid blood coagulation, and excellent wound closure in a rat model. ELISA study demonstrated the decreasing level of inflammatory markers, such as TNF-α, IL1β, and IL-6, making formulation promising for hemostatic wound healing applications. Finally, the study concludes that nanofibrous scaffolds loaded with TMB and VCM have promising potential as a dressing material for hemostatic wound healing applications.
The morbidity rate following a surgical procedure increasing rapidly in the cases associated with surgical site infections. Traditional sutures lack the ability to deliver drugs as the incorporation of the drug in their structure would hamper their mechanical properties. To prevent such infections, we developed an extracellular matrix mimicking electrospun nanofibrous yarns of poly-(D,L)-lactic acid and polyvinyl alcohol loaded with vancomycin and ferulic acid, prepared by uniaxial electrospinning technique. In-vitro characterization such as scanning electron microscopy, Fourier transform infrared spectroscopy, x-ray diffraction, tensile strength testing, degradation studies, and antimicrobial studies along with in-vivo evaluation done with help of incision wound healing rat model and simultaneous testing of microbial load in the incised tissue. The in-vitro studies indicated the nanofiber yarns have size range 200–300 nm with a tensile strength of 7.54 ± 0.58 MPa. The dual drug-loaded yarn showed sustained drug release over a period of 48 h. In-vitro water uptake and biodegradation data indicated optimum results suitable for suturing applications. Antimicrobial study showed excellent antimicrobial activity against both S. aureus and E. coli. Results obtained from in-vivo study suggested excellent wound healing potential of nanofiber yarns as compared with commercial silk sutures. The histopathological studies confirmed restoring ability of nanofiber yarn to the normal skin structure. Enzyme-linked immunosorbent assay (ELISA) study revealed the downregulation of inflammatory markers i.e. TNF-alpha and IL-6, making nanofibers sutures suitable for surgical wound healing applications. Overall, the present study may conclude that the developed dual drug-loaded nanofiber yarns have excellent potential in surgical wound healing applications.
To address the limitations of conventional therapies in psoriasis treatment, this study developed and evaluated a dual-drug-loaded nanofiber-based hydrogel film incorporating tazarotene (TZT) and calcipotriol (CPT). Electrospun nanofibers using a polyvinyl alcohol/polyvinylpyrrolidone (PVA/PVP) blend were embedded into a carbopol hydrogel matrix to improve drug delivery, skin permeability, and therapeutic efficacy. The optimized nanofibers exhibited uniform morphology (244–252 nm), favorable tensile strength (up to 22.5 MPa), and complete biodegradability within two weeks. Drug release studies confirmed sustained release (95.68% in 72 h) with Higuchi kinetics. In vitro and in vivo assessments—including MTT assay, antioxidant analysis, lipid profiling, and efficacy in an imiquimod-induced rat model—demonstrated superior antipsoriatic activity of the TZT–CPT nanofiber hydrogel over monotherapy systems. This multifunctional nanoplatform holds strong potential for effective psoriasis management through enhanced targeting, controlled release, and synergistic therapeutic action.
Electrospun composite nanofiber scaffolds are well known for their bone and tissue regeneration applications. This research is focused on the development of PVP and PVA nanofiber composite scaffolds enriched with hydroxyapatite (HA) nanoparticles and alendronate (ALN) using the electrospinning technique. The developed nanofiber scaffolds were investigated for their physicochemical as well as bone regeneration potential. The results obtained from particle size, zeta potential, SEM and EDX analysis of HA nanoparticles confirmed their successful fabrication. Further, SEM analysis verified nanofiber’s diameters within 200–250 nm, while EDX analysis confirmed the successful incorporation of HA and ALN into the scaffolds. XRD and TGA analysis revealed the amorphous and thermally stable nature of the nanofiber composite scaffolds. Contact angle, FTIR analysis, Swelling and biodegradability studies revealed the hydrophilicity, chemical compatibility, suitable water uptake capacity and increased in-vitro degradation making it appropriate for tissue regeneration. The addition of HA into nanofiber scaffolds enhanced the physiochemical properties. Additionally, hemolysis cell viability, cell adhesion and proliferation by SEM as well as confocal microscopy and live/dead assay results demonstrated the non-toxic and biocompatibility behavior of nanofiber scaffolds. Alkaline phosphatase (ALP) and tartrate-resistant acid phosphatase (TRAP) assays demonstrated osteoblast promotion and osteoclast inhibition, respectively. These findings suggest that developed HA and ALN-loaded PVP/PVA-ALN-HA nanofiber composite scaffolds hold significant promise for bone regeneration applications.
Electrospun composite nanofiber scaffolds are well known for their bone and tissue regeneration applications. This research is focused on the development of PVP and PVA nanofiber composite scaffolds enriched with hydroxyapatite (HA) nanoparticles and alendronate (ALN) using the electrospinning technique. The developed nanofiber scaffolds were investigated for their physicochemical as well as bone regeneration potential. The results obtained from particle size, zeta potential, SEM and EDX analysis of HA nanoparticles confirmed their successful fabrication. Further, SEM analysis verified nanofiber’s diameters within 200–250 nm, while EDX analysis confirmed the successful incorporation of HA and ALN into the scaffolds. XRD and TGA analysis revealed the amorphous and thermally stable nature of the nanofiber composite scaffolds. Contact angle, FTIR analysis, Swelling and biodegradability studies revealed the hydrophilicity, chemical compatibility, suitable water uptake capacity and increased in-vitro degradation making it appropriate for tissue regeneration. The addition of HA into nanofiber scaffolds enhanced the physiochemical properties. Additionally, hemolysis cell viability, cell adhesion and proliferation by SEM as well as confocal microscopy and live/dead assay results demonstrated the non-toxic and biocompatibility behavior of nanofiber scaffolds. Alkaline phosphatase (ALP) and tartrate-resistant acid phosphatase (TRAP) assays demonstrated osteoblast promotion and osteoclast inhibition, respectively. These findings suggest that developed HA and ALN-loaded PVP/PVA-ALN-HA nanofiber composite scaffolds hold significant promise for bone regeneration applications.
Hepatocellular carcinoma (HCC) is a common malignancy which affects a substantial number of individuals all over the globe. It is the third primary cause of death among persons with neoplasm and has the fifth largest mortality rate among men and the seventh highest mortality rate among women. Dalbergin (DL) is described to be effective in breast cancer via changing mRNA levels of apoptosis-related proteins. DL belongs to neoflavonoids, a drug category with low solubility and poor bioavailability. We created a synthetic version of this naturally occurring chemical, DL, and then analyzed it using 1H-NMR, 13C-NMR, and LC-MS. We also made PLGA nanoparticles and then coated them with galactose. The design of experiment software was used to optimize DL-loaded galactose-modified PLGA nanoparticles. The optimized DL-nanoformulations (DLF) and DL-modified nanoformulations (DLMF) were analyzed for particle size, polydispersity index, shape, and potential interactions. In-vitro experiments on liver cancer cell lines (HepG2) are used to validate the anti-proliferative efficacy of the modified DLMF. The in-vitro research on HepG2 cell lines also demonstrated cellular accumulation of DLF and DLMF by FITC level. The in-vitro result suggested that DLMF has high therapeutic effectiveness against HCC. In-vivo pharmacokinetics and bio-distribution experiments revealed that DLMF excelled pristine DL in terms of pharmacokinetic performance and targeted delivery, which is related to galactose’s targeting activity on the asialoglycoprotein receptor (ASGPR) in hepatic cells. Additionally, we performed an in-silico study of DL on caspase 3 and 9 proteins, and the results were found to be −6.7 kcal/mol and −6.6 kcal/mol, respectively. Our in-silico analysis revealed that the DL had strong apoptotic properties against HCC.
Diabetic foot ulceration is the most distressing complication of diabetes having no standard therapy. Nanofibers are an emerging and versatile nanotechnology-based drug-delivery system with unique wound-healing properties. This study aimed to prepare and evaluate silk-sericin based hybrid nanofibrous mats for diabetic foot ulcer. The nanofibrous mats were prepared by electrospinning using silk sericin mixed with different proportions of polycaprolactone (PCL) and cellulose acetate (CA) loaded with ferulic acid (FA). The in vitro characterizations, such as surface morphology, mechanical properties, swelling behavior, biodegradability, scanning electron microscopy, and drug release were carried out. The SEM images indicated that nanofibers formed with varied diameters, ranging from 100 to 250 nm, and their tensile strength was found to range from 7 to 15 MPa. In vitro release demonstrated that the nanofibers sustained FA release over an extended time of period. In vitro cytotoxicity showed that the nanofibers possessed a lower cytotoxicity in HaCaT cells. The in vivo wound-healing studies demonstrated an excellent wound-healing efficiency of the nanofibers in diabetic rats. Furthermore, the histopathological studies showed the nanofibers’ ability to restore the skin’s normal structure. Therefore, it was concluded that the prepared silk-sericin-based hybrid nanofibers loaded with FA could be a promising drug-delivery platform for the effective treatment of diabetic foot ulcers.
Diabetes mellitus is a chronic disease with a high mortality rate and many complications. A non-healing diabetic foot ulcer (DFU) is one the most serious complications, leading to lower-extremity amputation in 15% of diabetic patients. Nanofibers are emerging as versatile wound dressing due to their unique wound healing properties, such as a high surface area to volume ratio, porosity, and ability to maintain a moist wound environment capable of delivering sustained drug release and oxygen supply to a wound. The present study was aimed to prepare and evaluate a polyvinyl alcohol (PVA)–sodium alginate (SA)–silk fibroin (SF)-based multifunctional nanofibrous scaffold loaded with asiaticoside (AT) in diabetic rats. The SEM findings showed that fibers’ diameters ranged from 100–200 nm, and tensile strengths ranged from 12.41–16.80 MPa. The crosslinked nanofibers were sustained AT over an extended period. The MTT and scratch assay on HaCat cells confirmed low cytotoxicity and significant cell migration, respectively. Antimicrobial tests revealed an excellent anti-microbial efficacy against P. aeruginosa and S. aureus bacteria. In-vivo study demonstrated better wound healing efficacy in diabetic rats. In addition, the histopathological studies showed its ability to restore the normal structure of the skin. The present study concluded that developed multifunctional nanofibers have a great potential for diabetic wound healing applications.
This study focuses on the fabrication and evaluation of a silk fibroin–based electrospun nanofiber system co-formulated with curcumin, polycaprolactone (PCL), and polyvinyl alcohol (PVA) to enhance diabetic wound healing. Silk fibroin, a natural biopolymer, was selected for its biodegradability, oxygen permeability, and moisture retention—critical factors in effective wound management. The nanofibers exhibited uniform morphology with diameters ranging from 200–350 nm and tensile strength between 12.41–16.80 MPa. Sustained curcumin release was achieved, enhancing its therapeutic window. In vivo studies in streptozotocin-induced diabetic mice demonstrated significantly improved wound closure and tissue regeneration compared to conventional formulations. Histopathological analysis confirmed restored skin architecture and reduced inflammation. The synergistic combination of silk fibroin and curcumin provides antioxidant and anti-inflammatory benefits, making this nanofiber dressing a promising candidate for diabetic wound therapy.