1
|
Moghimipour E, Khazali M, MakhmalZadeh BS, Abedini Baghbadorani M, Zangeneh A, Sohrabi S, Nejaddehbashi F, Hajipour F, Handali S. Development of propranolol loaded SLN for transdermal delivery: in-vitro characterization and skin deposition studies. Ther Deliv 2025; 16:205-215. [PMID: 39874079 PMCID: PMC11875500 DOI: 10.1080/20415990.2025.2458451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 01/22/2025] [Indexed: 01/30/2025] Open
Abstract
AIM The study aimed to formulate solid lipid nanoparticles (SLNs) for the transdermal delivery of PPL to improve skin retention and efficacy. MATERIALS AND METHOD The particle size distribution of SLNs was determined and the morphology of SLNs was also analyzed by SEM. In-vitro, ex-vivo and in vivo evaluations were done for PPL loaded SLN. The safety of drug delivery systems was assayed using MTT test. RESULTS The results indicated successful encapsulation of PPL in SLNs (59.38%), which exhibited a spherical shape and smooth surface. Compared to PPL solution, SLNs demonstrated a prolonged drug release profile in vitro. Stability tests over three months showed no significant changes in entrapment efficiency or size distribution. Enhanced permeation through shed snake and rat skin was observed with SLNs compared to the PPL solution. Ex-vivo and in vivo studies confirmed that PPL-loaded SLNs significantly increased drug content in the skin. Importantly, the SLNs displayed biocompatibility, as no significant cytotoxic effects were noted, and they were nonirritating to rat skin. CONCLUSION To the best of our knowledge, this is the first study that indicates SLNs can be considered as a promising nanocarriers for transdermal delivery of PPL.
Collapse
Affiliation(s)
- Eskandar Moghimipour
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammadamin Khazali
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | | | - Ali Zangeneh
- Nanotechnology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Somayeh Sohrabi
- Mechanical Engineering Department, Sharif University of Technology, Tehran, Iran
| | - Fereshteh Nejaddehbashi
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Hajipour
- Department of Medicinal Chemistry, Faculty of pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Handali
- Medical Biomaterials Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Aboud HM, Ali AA, Mohammed NH, Hassan AHE, Roh EJ, El Menshawe SF. Investigating the Potential of Ufasomes Laden with Nintedanib as an Optimized Targeted Lung Nanoparadigm for Accentuated Tackling of Idiopathic Pulmonary Fibrosis. Pharmaceuticals (Basel) 2024; 17:1605. [PMID: 39770447 PMCID: PMC11677946 DOI: 10.3390/ph17121605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/11/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
Background/objectives: Idiopathic pulmonary fibrosis (IPF) is a prevalent interstitial lung disease that typically progresses gradually, leading to respiratory failure and ultimately death. IPF can be treated with the tyrosine kinase inhibitor, nintedanib (NTD), owing to its anti-fibrotic properties, which ameliorate the impairment of lung function. This study aimed to formulate, optimize, and assess NTD-loaded ufasomes (NTD-UFSs) as a nanosystem for its pulmonary targeting to snowball the bioavailability and therapeutic efficacy of the drug. Methods: To investigate the influence of numerous factors on NTD-UFSs assembly and to determine the optimal formulation, Box-Behnken statistical design was implemented with the assistance of Design-Expert® software. The thin-film hydration strategy was employed to fabricate NTD-UFSs. The optimum NTD-UFSs formulation was subsequently selected and subjected to additional evaluations. Also, using a rat model, a comparative pharmacokinetic analysis was scrutinized. Results: The optimal NTD-UFSs elicited an accumulative release of 65.57% after 24 h, an encapsulation efficiency of 62.51%, a zeta potential of -36.07 mV, and a vesicular size of 364.62 nm. In addition, it disclosed remarkable stability and a continuous cumulative release pattern. In vivo histopathological studies ascertained the tolerability of NTD-UFSs administered intratracheally. According to the pharmacokinetic studies, intratracheal NTD-UFSs administration manifested a significantly higher AUC0-∞ value than oral and intratracheal NTD suspensions, by approximately 5.66- and 3.53-fold, respectively. Conclusions: The findings of this study proposed that UFSs might be a promising nanoparadigm for the non-invasive pulmonary delivery of NTD.
Collapse
Affiliation(s)
- Heba M. Aboud
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt; (H.M.A.); (A.A.A.); (S.F.E.M.)
| | - Adel A. Ali
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt; (H.M.A.); (A.A.A.); (S.F.E.M.)
| | - Nada H. Mohammed
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Deraya University, Minia 61768, Egypt
| | - Ahmed H. E. Hassan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Eun Joo Roh
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Shahira F. El Menshawe
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt; (H.M.A.); (A.A.A.); (S.F.E.M.)
| |
Collapse
|
3
|
Aati S, Farouk HO, Elkarmalawy MH, Aati HY, Tolba NS, Hassan HM, Rateb ME, Hamad DS. Intratracheal Administration of Itraconazole-Loaded Hyaluronated Glycerosomes as a Promising Nanoplatform for the Treatment of Lung Cancer: Formulation, Physiochemical, and In Vivo Distribution. Pharmaceutics 2024; 16:1432. [PMID: 39598555 PMCID: PMC11597389 DOI: 10.3390/pharmaceutics16111432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Itraconazole (ITZ) is an antiangiogenic agent recognized as a potent suppressor of endothelial cell growth that suppresses angiogenesis. Nevertheless, its exploitation is significantly restricted by its low bioavailability and systematic side effects. The objective of this study was to utilize glycerosomes (GLY), glycerol-developed vesicles, as innovative nanovesicles for successful ITZ pulmonary drug delivery. METHODS The glycerosomes were functionalized with hyaluronic acid (HA-GLY) to potentiate the anticancer efficacy of ITZ and extend its local bio-fate. ITZ-HA-GLY were fabricated using soybean phosphatidylcholine, tween 80, HA, and sonication time via a thin-film hydration approach according to a 24 full factorial design. The impact of formulation parameters on ITZ-HA-GLY physicochemical properties, as well as the optimal formulation option, was evaluated using Design-Expert®. Sulphorhodamine-B (SRB) colorimetric cytotoxicity assay of the optimized ITZ-HA-GLY versus ITZ suspension was explored in the human A549 cell line. The in vivo pharmacokinetics and bio-distribution examined subsequent to intratracheal administrations of ITZ suspension, and ITZ-HA-GLY were scrutinized in rats. RESULTS The optimized ITZ-HA-GLY unveiled vesicles of size 210.23 ± 6.43 nm, zeta potential of 41.06 ± 2.62 mV, and entrapment efficiency of 73.65 ± 1.76%. Additionally, ITZ-HA-GLY manifested a far lower IC50 of 13.03 ± 0.2 µg/mL on the A549 cell line than that of ITZ suspension (28.14 ± 1.6 µg/mL). Additionally, the biodistribution analysis revealed a higher concentration of ITZ-HA-GLY within the lung tissues by 3.64-fold as compared to ITZ suspension. Furthermore, the mean resistance time of ITZ-HA-GLY declined more slowly with 14 h as compared to ITZ suspension, confirming the accumulation of ITZ inside the lungs and their promising usage as a target for the treatment of lung disease. CONCLUSIONS These data indicate that the improved ITZ-HA-GLY demonstrates significant promise and represents an exciting prospect in intratracheal delivery systems for lung cancer treatment, meriting further investigation.
Collapse
Affiliation(s)
- Sultan Aati
- Dental Health Department, College of Applied Medical Sciences, King Saud University, Riyadh 11421, Saudi Arabia;
| | - Hanan O. Farouk
- Department of Pharmaceutics, Faculty of Pharmacy, Nahda University, Beni-Suef 62521, Egypt;
| | - Marwa H. Elkarmalawy
- Department of Pharmaceutics and Drug Manufacturing, Faculty of Pharmacy, Modern University for Technology and Information, Cairo 11571, Egypt;
| | - Hanan Y. Aati
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
| | - Nahla Sameh Tolba
- Department of Pharmaceutics, Faculty of Pharmacy, Sadat City University, Sadat City 32897, Egypt;
| | - Hossam M. Hassan
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62521, Egypt
| | - Mostafa E. Rateb
- School of Computing, Engineering & Physical Sciences, University of the West of Scotland, Paisley PA1 2BE, UK;
| | - Doaa S. Hamad
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Nile Valley University, Fayoum 63518, Egypt;
| |
Collapse
|
4
|
Heikal LA, Ashour AA, Aboushanab AR, El-Kamel AH, Zaki II, El-Moslemany RM. Microneedles integrated with atorvastatin-loaded pumpkisomes for breast cancer therapy: A localized delivery approach. J Control Release 2024; 376:354-368. [PMID: 39413849 DOI: 10.1016/j.jconrel.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/20/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
Breast cancer is the most common invasive cancer in women worldwide, having a significant impact on women's well-being. Early diagnosis of breast cancer followed by appropriate treatment is considered the best survival factor. Microneedles (MN) have been utilized for non-invasive localized breast cancer treatment. The combination of nano-carriers with MN technology represents an appealing strategy for improving drug delivery efficacy. It is worth noting that atorvastatin (ATV) has received substantial interest as a drug with potential anticancer activity. Our study aimed to formulate an ATV-loaded bioactive pumpkin seed oil vesicular nanocarrier; pumpkisomes (PUMP) for enhanced localized delivery to breast cancer using MN. The selected PUMP formulation had a particle size of 151.8 ± 2.7 nm, zeta potential of -54.1 mV, and % entrapment efficiency of 73 %. PUMP showed a sustained ATV release, potent selective cytotoxic effect (IC50 of 2.82 ± 0.02 μg/mL), enhanced internalization (2.8-fold increase compared to the free drug), and potent anti-migratory effect on MDA-MB-231 cells (21.15 ± 3.6 % wound closure compared to 80.81 ± 4.1 % for free drug). Moreover, integrating ATV-PUMP in dissolving microneedles (ATV-PUMP@dMN) showed a quick dissolution rate and appropriate mechanical strength with high piercing efficiency. ATV permeation across the skin from ATV-PUMP@dMN was also improved (1.8-fold increase compared to ATV-PUMP@gel). ATV-PUMP@dMN demonstrated an efficient anticancer effect when applied in an Ehrlich ascites mammary tumor model attaining significant improvement in ATV antiproliferative (PTEN and Ki-67), antiangiogenic (VEGF) and apoptotic (Bcl2, Bax and caspase3) effects restoring tumor biomarkers to levels comparable to the negative control group. Thus, our study presents PUMP as a novel and promising bioactive vesicular nanosystem with potential synergistic effect with ATV or other antitumor drugs. PUMP-integrated MN could be considered a promising platform for future applications in localized breast cancer therapy.
Collapse
Affiliation(s)
- Lamia A Heikal
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| | - Asmaa A Ashour
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Alaa R Aboushanab
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Amal H El-Kamel
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Inass I Zaki
- Department of Pathology, Faculty of Medicine Alexandria University, Alexandria, Egypt
| | - Riham M El-Moslemany
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
5
|
Salem HF, Nafady MM, Khallaf RA, Abdel-Sattar AR, Abdel-Sattar HH, Eissa EM. Implementing losartan potassium-laden pegylated nanocubic vesicles as a novel nanoplatform to alleviate cisplatin-induced nephrotoxicity via blocking apoptosis and activating the wnt/β-catenin/TCF-4 pathway. Life Sci 2024; 354:122955. [PMID: 39122109 DOI: 10.1016/j.lfs.2024.122955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/16/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
AIMS Losartan potassium-laden pegylated nanocubic vesicles (LP-NCVs-PEG) have an intriguing kidney-targeted nanoplatform for acute renal injury via blocking apoptosis and activating wnt/β-catenin pathway. MAIN METHODS Utilizing a thin-film hydration methodology established on 42 full factorial design to produce LP loaded nanocubic formulations (LP-NCVs) which composed mainly from L-α-phosphatidylcholine and poloxamer. The optimization process was designed to select the formulation with maximum entrapment efficiency (EE %), maximum in-vitro drug release (Q8h), and minimum vesicle size (VS). The optimum formulation was then pegylated to obtain LP-NCVs-PEG formulation that shields NCVs from the harsh ecosystem of the stomach, improves their oral drug delivery performance and targets the proximal renal tubules with no systemic toxicity. Male albino rats were injected with Cisplatin (6 mg/kg, i.p.) alone or with LP-formulations (5 mg/kg/day). Kidney injury markers, inflammatory markers, apoptotic markers. Besides renal tissue expression of Wnt, β-Catenin, GSK-3β, renal RNA gene expression of TCF-4, LEF-1 and histopathology were also analyzed to display pharmacological study. KEY FINDINGS The pharmacokinetics studies demonstrated that LP-NCVs-PEG boosted LP bioavailability approximately 3.61 times compared to LP oral solution. Besides LP-NCVs-PEG may have an intriguing kidney-targeted nanoplatform for acute renal injury via decreased renal toxicity markers, renal expression of LEF-1, GSK3-β, caspase, TNF-α, NF-κB and TUNEL expression. Alternatively, increased renal tissue level of Bcl-2, wnt, β-catenin and TCF-4. SIGNIFICANCE LP-NCVs-PEG improved LP pharmacokinetics targeting the kidney and improved injury by activating wnt/β-catenin/TCF-4 pathway, blocking apoptosis, inflammation and renal toxicity markers suggesting it might be successful nephroprotective adjuvant therapy.
Collapse
Affiliation(s)
- Heba F Salem
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.
| | - Mohamed M Nafady
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Nahda University Beni-Suef, Egypt.
| | - Rasha A Khallaf
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.
| | | | - Hend Hassan Abdel-Sattar
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Nahda University Beni-Suef, Egypt.
| | - Essam M Eissa
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.
| |
Collapse
|
6
|
Salem HF, Nafady MM, Eissa EM, Abdel-Sattar HH, Khallaf RA. Assembly of In-Situ Gel Containing Nano-Spanlastics of an Angiotensin II Inhibitor as a Novel Epitome for Hypertension Management: Factorial Design Optimization, In-vitro Gauging, Pharmacokinetics, and Pharmacodynamics Appraisal. AAPS PharmSciTech 2024; 25:115. [PMID: 38755324 DOI: 10.1208/s12249-024-02823-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/29/2024] [Indexed: 05/18/2024] Open
Abstract
More than 1 billion people worldwide suffer from hypertension; therefore, hypertension management has been categorized as a global health priority. Losartan potassium (LP) is an antihypertensive drug with a limited oral bioavailability of about 33% since it undergoes the initial metabolic cycle. Thus, nasal administration is a unique route to overcome first-pass metabolism. The investigation focused on the potential effects of LP-loaded spanlastic vesicles (SNVs) on LP pharmacodynamics and pharmacokinetic parameters, utilizing a thin-film hydration methodology established on a 3122 full factorial design. Entrapment efficiency (EE%) ranged from 39.8 ± 3.87.8 to 83.8 ± 2.92% for LP-SNVs. Vesicle size (VS) varied from 205.5 ± 6.5.10 to 445.1 ± 13.52 nm, and the percentage of LP released after 8 h (Q8h) ranged from 30.8 ± 3.10 to 68.8 ± 1.45%. LP permeated through the nasal mucosa during 24 h and flocculated from 194.1 ± 4.90 to 435.3 ± 13.53 µg/cm2. After twenty-four hours, the optimal LP-SNVs in-situ gel showed 2.35 times more permeation through the nasal mucosa than the LP solution. It also lowered systolic blood pressure, so it is thought to be better than the reference formulation in terms of pharmacodynamics. The pharmacokinetics studies demonstrated that the intranasal LP-SNVs gel boosted its bioavailability approximately 6.36 times compared to the oral LP solution. Our research showed that intranasal LP-SNVs could be a good nanoplatform because they are well-tolerated and have possible pharmacokinetics and pharmacodynamics.
Collapse
Affiliation(s)
- Heba F Salem
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed M Nafady
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| | - Essam M Eissa
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Hend Hassan Abdel-Sattar
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| | - Rasha A Khallaf
- Pharmaceutics and Industrial Pharmacy Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.
| |
Collapse
|
7
|
Park SY, Jung JH, Kim DS, Lee JK, Song BG, Shin HE, Jung JW, Baek SW, You S, Han I, Han DK. Therapeutic potential of luteolin-loaded poly(lactic-co-glycolic acid)/modified magnesium hydroxide microsphere in functional thermosensitive hydrogel for treating neuropathic pain. J Tissue Eng 2024; 15:20417314231226105. [PMID: 38333057 PMCID: PMC10851718 DOI: 10.1177/20417314231226105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 12/27/2023] [Indexed: 02/10/2024] Open
Abstract
Neuropathic pain (NP) is a debilitating condition stemming from damage to the somatosensory system frequently caused by nerve injuries or lesions. While existing treatments are widely employed, they often lead to side effects and lack specificity. This study aimed to alleviate NP by developing an innovative sustained-release thermosensitive hydrogel system. The system incorporates hyaluronic acid (HA)/Pluronic F127 injectable hydrogel and bupivacaine (Bup, B) in combination with poly(lactic-co-glycolic acid; PLGA)/modified magnesium hydroxide (MH)/luteolin (Lut; PML) microspheres (PML@B/Gel). The PML@B/Gel was designed for localized and prolonged co-delivery of Bup and Lut as an anesthetic and anti-inflammatory agent, respectively. Our studies demonstrated that PML@B/Gel had exceptional biocompatibility, anti-inflammatory, and antioxidant properties. In addition, it exhibited efficient pain relief in in vitro cellular assays. Moreover, this functional hydrogel showed substantial sustained drug release while diminishing microglial activation. Consequently, it effectively mitigated mechanical allodynia and thermal hyperalgesia in in vivo rat models of chronic constriction injury (CCI). Based on our research findings, PML@B/Gel emerges as a promising therapeutic approach for the protracted treatment of NP.
Collapse
Affiliation(s)
- So-Yeon Park
- Department of Biomedical Science, CHA University, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, Korea
| | - Joon Hyuk Jung
- Department of Life Science, CHA University School of Medicine, Seongnam-si, Gyeonggi-do, Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, USA
| | - Jun-Kyu Lee
- Department of Biomedical Science, CHA University, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
| | - Byeong Gwan Song
- Department of Life Science, CHA University School of Medicine, Seongnam-si, Gyeonggi-do, Korea
| | - Hae Eun Shin
- Department of Life Science, CHA University School of Medicine, Seongnam-si, Gyeonggi-do, Korea
| | - Ji-Won Jung
- Department of Biomedical Science, CHA University, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
| | - Seung-Woon Baek
- Department of Biomedical Science, CHA University, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
| | - Seungkwon You
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, Korea
| | - Inbo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
| |
Collapse
|
8
|
Yalçın MB, Bora ES, Erdoğan MA, Çakır A, Erbaş O. The Effect of Adipose-Derived Mesenchymal Stem Cells on Peripheral Nerve Damage in a Rodent Model. J Clin Med 2023; 12:6411. [PMID: 37835055 PMCID: PMC10573691 DOI: 10.3390/jcm12196411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/27/2023] [Accepted: 10/07/2023] [Indexed: 10/15/2023] Open
Abstract
Peripheral nerve damage is a significant clinical problem with limited therapeutic options. Adipose-derived mesenchymal stem cells (ADSCs) have emerged as a promising therapeutic approach due to their regenerative potential. However, the underlying mechanisms by which ADSCs promote peripheral nerve regeneration remain unclear. In this study, we investigated the role of syndecan-1 and heat shock protein 70 (HSP-70) in mediating the regenerative effects of ADSCs on peripheral nerves. ADSCs were characterized and isolated from the adipose tissue of rats. In vitro experiments were conducted to evaluate the ability of ADSCs to secrete syndecan-1 and HSP-70 in response to stress conditions. To evaluate the therapeutic potential of ADSCs, rats with sciatic nerve injuries were treated with ADSCs and assessed for functional recovery, nerve regeneration, and changes in syndecan-1 and HSP-70 levels. Regeneration was evaluated with Electromyography (EMG) histology. The results showed that ADSCs could secrete syndecan-1 and HSP-70 in response to stress conditions. Furthermore, ADSC treatment significantly improved functional recovery and nerve regeneration and increased syndecan-1 and HSP-70 levels in the injured nerve. On the other hand, ADSCs make improvements histologically through the influence of Nerve growth factor (NGF), Malondialdehyde (MDA), and EMG.
Collapse
Affiliation(s)
- Mehmet Burak Yalçın
- Department of Orthopedics and Traumatology, Bahcelievler Memorial Hospital, Istanbul 34180, Turkey;
| | - Ejder Saylav Bora
- Department of Emergency Medicine, Izmir Atatürk Research and Training Hospital, Izmir 35360, Turkey
| | - Mümin Alper Erdoğan
- Department of Physiology, Faculty of Medicine, Izmir Kâtip Çelebi University, Izmir 35620, Turkey;
| | - Adem Çakır
- Department of Emergency Medicine, Çanakkale Mehmet Akif Ersoy State Hospital, Çanakkale 17100, Turkey;
| | - Oytun Erbaş
- Department of Physiology, Demiroğlu Bilim University, Istanbul 34394, Turkey;
| |
Collapse
|
9
|
Novel Bile Salt Stabilized Vesicles-Mediated Effective Topical Delivery of Diclofenac Sodium: A New Therapeutic Approach for Pain and Inflammation. Pharmaceuticals (Basel) 2022; 15:ph15091106. [PMID: 36145327 PMCID: PMC9506322 DOI: 10.3390/ph15091106] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
The oral delivery of diclofenac sodium (DNa), a non-steroidal analgesic, anti-inflammatory drug, is associated with various gastrointestinal side effects. The aim of the research was to appraise the potential of transdermal delivery of DNa using bilosomes as a vesicular carrier (BSVC) in inflamed paw edema. DNa-BSVCs were elaborated using a thin-film hydration technique and optimized using a 31.22 multilevel categoric design with Design Expert® software 10 software (Stat-Ease, Inc., Minneapolis, MI, USA). The effect of formulation variables on the physicochemical properties of BSVC, as well as the optimal formulation selection, was investigated. The BSVCs were evaluated for various parameters including entrapment efficiency (EE%), vesicle size (VS), zeta potential (ZP) and permeation studies. The optimized BSVC was characterized for in vitro release, Fourier transform infrared spectroscopy (FTIR), transmission electron microscopy (TEM) and incorporated into hydrogel base. The optimized DNa-BSVC gel effectiveness was assessed in vivo using carrageenan-induced paw edema animal model via cyclooxygenase 2 (COX-2), interleukin 6 (IL-6), Hemooxygenase 1 (HO-1) and nuclear factor-erythroid factor2-related factor 2 (Nfr-2) that potentiate anti-inflammatory and anti-oxidant activity coupled with histopathological investigation. The resulting vesicles presented VS from 120.4 ± 0.65 to 780.4 ± 0.99 nm, EE% from 61.7 ± 3.44 to 93.2 ± 2.21%, ZP from −23.8 ± 2.65 to −82.1 ± 12.63 mV and permeation from 582.9 ± 32.14 to 1350.2 ± 45.41 µg/cm2. The optimized BSVCs were nano-scaled spherical vesicles with non-overlapped bands of their constituents in the FTIR. Optimized formulation has superior skin permeability ex vivo approximately 2.5 times greater than DNa solution. Furthermore, histological investigation discovered that the formed BSVC had no skin irritating properties. It was found that DNa-BSVC gel suppressed changes in oxidative inflammatory mediators (COX-2), IL-6 and consequently enhanced Nrf2 and HO-1 levels. Moreover, reduction of percent of paw edema by about three-folds confirmed histopathological alterations. The results revealed that the optimized DNa-BSVC could be a promising transdermal drug delivery system to boost anti-inflammatory efficacy of DNa by enhancing the skin permeation of DNa and suppressing the inflammation of rat paw edema.
Collapse
|