1
|
Sadeghi-Avalshahr A, Nazarnezhad S, Hassanzadeh H, Kazemi Noughabi M, Namaei-Ghasemnia N, Jalali M. Synergistic effects of incorporated additives in multifunctional dressings for chronic wound healing: An updated comprehensive review. Wound Repair Regen 2025; 33:e13238. [PMID: 39682073 DOI: 10.1111/wrr.13238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024]
Abstract
Detailed reviewing of the complicated process of wound healing reveals that it resembles an orchestrated symphony via a precise and calculated collaboration of relevant cells at the wound site. The domino-like function of various cytokines, chemokines, growth factors and small biological molecules such as antibacterial peptides all come together to successfully execute the wound healing process. Therefore, it appears that the use of a wound dressing containing only a single additive with specific properties and capabilities may not be particularly effective in treating the complex conditions that are usual in the environment of chronic wounds. The use of multifunctional dressings incorporating various additives has shown promising results in enhancing wound healing processes. This comprehensive review article explores the synergistic effects of integrated additives in such dressings, aiming to provide an updated understanding of their combined therapeutic potential. By analysing recent advancements and research findings, this review sheds light on the intricate interactions between different additives, their mechanisms of action and their cumulative impact on wound healing outcomes. Moreover, the review discusses the importance of utilising combined therapies in wound care and highlights the potential future directions and implications for research and clinical practice in the field of wound healing management.
Collapse
Affiliation(s)
- Alireza Sadeghi-Avalshahr
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Materials Research, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Simin Nazarnezhad
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Halimeh Hassanzadeh
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture, and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| | - Mahboubeh Kazemi Noughabi
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture, and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| | - Negar Namaei-Ghasemnia
- Department of Materials Research, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Mehdi Jalali
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
2
|
Lu C, Wu X, Meng X, Liu Y, Yang T, Zeng Y, Chen Y, Huang Y, Fang Z, Yang X, Luo J. Silver Nanoparticles Exposure Impairs Cardiac Development by Suppressing the Focal Adhesion Pathway in Zebrafish. Int J Nanomedicine 2024; 19:9291-9304. [PMID: 39282573 PMCID: PMC11400637 DOI: 10.2147/ijn.s476168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction The potential toxic effects of wastewater discharges containing silver nanoparticles (AgNPs) and their release into aquatic ecosystems on aquatic organisms are becoming a major concern for environmental and human health. However, the potential risks of AgNPs to aquatic organisms, especially for cardiac development by Focal adhesion pathway, are still poorly understood. Methods The cardiac development of various concentrations of AgNPs in zebrafish were examined using stereoscopic microscope. The expression levels of cardiac development-related genes were analyzed by qRT-PCR and Whole-mount in situ hybridization (WISH). In addition, Illumina high-throughput global transcriptome analysis was performed to explore the potential signaling pathway involved in the treatment of zebrafish embryos by AgNPs after 72 h. Results We systematically investigated the cardiac developing toxicity of AgNPs on the embryos of zebrafish. The results demonstrated that 2 or 4 mg/L AgNPs exposure induces cardiac developmental malformations, such as the appearance of pericardial edema phenotype. In addition, after 72 h of exposure, the mRNA levels of cardiac development-related genes, such as myh7, myh6, tpm1, nppa, tbx5, tbx20, myl7 and cmlc1, were significantly lower in AgNPs-treated zebrafish embryos than in control zebrafish embryos. Moreover, RNA sequencing, KEGG (Kyoto Encyclopedia of Genes) and Genomes and GSEA (gene set enrichment analysis) of the DEGs (differentially expressed genes) between the AgNPs-exposed and control groups indicated that the downregulated DEGs were mainly enriched in focal adhesion pathways. Further investigations demonstrated that the mRNA levels of focal adhesion pathway-related genes, such as igf1ra, shc3, grb2b, ptk2aa, akt1, itga4, parvaa, akt3b and vcla, were significantly decreased after AgNPs treatment in zebrafish. Conclusion Thus, our findings illustrated that AgNPs could impair cardiac development by regulating the focal adhesion pathway in zebrafish.
Collapse
Affiliation(s)
- Chunjiao Lu
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Xuewei Wu
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Xin Meng
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Yi Liu
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Ting Yang
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Yan Zeng
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Yang Chen
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Yishan Huang
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Zhou Fang
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Xiaojun Yang
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Juanjuan Luo
- Engineering Research Center of Key Technique for Biotherapy of Guangdong Province, Shantou University Medical College, Shantou, 515041, People's Republic of China
| |
Collapse
|
3
|
Fayyazi F, Ebrahimi V, Mamaghani MM, Abgharmi BA, Zarrini G, Mosarrezaii A, Charkhian H, Gholinejad Z. N-Acetyl cysteine amide and cerium oxide nanoparticles as a drug delivery for ischemic stroke treatment: Inflammation and oxidative stress crosstalk. J Trace Elem Med Biol 2023; 80:127300. [PMID: 37741051 DOI: 10.1016/j.jtemb.2023.127300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/25/2023]
Abstract
BACKGROUND Inflammation and oxidative stress crosstalk is involved in the ischemic stroke(IS) pathogenesis and the new therapeutic options should be offered based on the targets that are critical in the golden hour of IS. YKL-40 and total antioxidant capacity(TAC), the inflammation and oxidative stress biomarkers, provide us with clues for proper intervention targets. N-acetyl cysteine amide (NACA), a lipophilic antioxidant, with a nanoparticle-based drug delivery system is permeable enough to penetrate blood-brain barrier (BBB) and was proposed as a new treatment option for IS. In this study, we evaluated the YKL-40 and TAC levels in the sera of IS patients to elucidate the best intervention target. A rat tissue model is used to assess the NACA efficiency. The microbiology tests performed to figure out the potential NACA and antibiotics interactions. MATERIAL AND METHODS The YKL-40 and TAC were measured in the serum of IS patients by ELISA and FRAP methods, respectively. The serum samples were obtained 12 h after the patient's admission and meantime other laboratory findings and NIHSS-based prognosis were recorded. In the animal study, the brain cortex, liver, kidney, adipose, and the heart of healthy rats were dissected and then incubated in DMEM cell culture media containing 50 micrograms/milliliter of nanoparticles; the nanoparticles were titanium dioxide nanoparticles (TiO2 NPs), copper oxide nanoparticles (CuO NPs) and cerium dioxide nanoparticles (CeO2 NPs). Olive oil and human serum albumin solution were exposed to the nanoparticles with and without NACA. TAC was measured in the supernatant culture media. With similar concentrations and settings, we evaluated the NACA, nanoparticle, and antibiotics interactions on pseudomonas aeruginosa. RESULTS There was a nonparametric correlation between YKL-40 levels and post stroke serum TAC levels. Nonsmokers had higher YKL-40 and TAC levels than smokers. A new calculated variable, urea*lymphocyte/age, predicts a poor prognosis with an acceptable AUC (0.708). Exposing to the nanoparticles, the liver, kidney, and brain had a significantly higher TAC than adipose and cardiac tissue. The NACA had an ameliorative effect against TiO2 NPs in the brain. This effectiveness of NACA was also observed against CuO NPs treatment. However, the CeO2 NPs exert a strong antioxidant property by reducing the TAC in the brain tissue but not the others. Albumin showed antioxidant properties by itself, but olive oil had an inert behavior. NACA had no interaction with the action of routine antibiotics. CONCLUSION Oxidative stress but not inflammation is the best point for intervention in IS patients because YKL-40 has not a relationship with NIHSS score. The CeO2 NPs and NACA combination are eligible option to develop antioxidant-based drug for the treatment of IS. As a complementary finding, the urea*lymphocyte/age is proposed as a NIHSS-based prognosis biomarker.
Collapse
Affiliation(s)
- Farzin Fayyazi
- Department of Neurology, Urmia University of Medical Sciences, Urmia, Iran
| | - Vahed Ebrahimi
- Department of Biochemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Benyamin Azad Abgharmi
- Department of Microbiology Science, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Gholamreza Zarrini
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Arash Mosarrezaii
- Department of Neurology, Urmia University of Medical Sciences, Urmia, Iran.
| | - Hamed Charkhian
- Young Researchers and Elite Club, Islamic Azad University, Urmia Branch, Urmia, Iran
| | - Zafar Gholinejad
- Department of Medical Laboratory Science, Urmia Branch, Islamic Azad University, Urmia, Iran.
| |
Collapse
|
4
|
Roshani M, Rezaian-Isfahni A, Lotfalizadeh MH, Khassafi N, Abadi MHJN, Nejati M. Metal nanoparticles as a potential technique for the diagnosis and treatment of gastrointestinal cancer: a comprehensive review. Cancer Cell Int 2023; 23:280. [PMID: 37981671 PMCID: PMC10657605 DOI: 10.1186/s12935-023-03115-1] [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: 04/11/2023] [Accepted: 10/27/2023] [Indexed: 11/21/2023] Open
Abstract
Gastrointestinal (GI) cancer is a major health problem worldwide, and current diagnostic and therapeutic approaches are often inadequate. Various metallic nanoparticles (MNPs) have been widely studied for several biomedical applications, including cancer. They may potentially overcome the challenges associated with conventional chemotherapy and significantly impact the overall survival of GI cancer patients. Functionalized MNPs with targeted ligands provide more efficient localization of tumor energy deposition, better solubility and stability, and specific targeting properties. In addition to enhanced therapeutic efficacy, MNPs are also a diagnostic tool for molecular imaging of malignant lesions, enabling non-invasive imaging or detection of tumor-specific or tumor-associated antigens. MNP-based therapeutic systems enable simultaneous stability and solubility of encapsulated drugs and regulate the delivery of therapeutic agents directly to tumor cells, which improves therapeutic efficacy and minimizes drug toxicity and leakage into normal cells. However, metal nanoparticles have been shown to have a cytotoxic effect on cells in vitro. This can be a concern when using metal nanoparticles for cancer treatment, as they may also kill healthy cells in addition to cancer cells. In this review, we provide an overview of the current state of the field, including preparation methods of MNPs, clinical applications, and advances in their use in targeted GI cancer therapy, as well as the advantages and limitations of using metal nanoparticles for the diagnosis and treatment of gastrointestinal cancer such as potential toxicity. We also discuss potential future directions and areas for further research, including the development of novel MNP-based approaches and the optimization of existing approaches.
Collapse
Affiliation(s)
- Mohammad Roshani
- Internal Medicine and Gastroenterology, Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Arya Rezaian-Isfahni
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Negar Khassafi
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Hassan Jafari Najaf Abadi
- Research Center for Health Technology Assessment and Medical Informatics, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Majid Nejati
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
5
|
Rybka M, Mazurek Ł, Konop M. Beneficial Effect of Wound Dressings Containing Silver and Silver Nanoparticles in Wound Healing-From Experimental Studies to Clinical Practice. LIFE (BASEL, SWITZERLAND) 2022; 13:life13010069. [PMID: 36676019 PMCID: PMC9864212 DOI: 10.3390/life13010069] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Impaired wound healing affects hundreds of million people around the world; therefore, chronic wounds are a major problem not only for the patient, but also for already overloaded healthcare systems. Chronic wounds are always very susceptible to infections. Billions of dollars are spent to discover new antibiotics as quickly as possible; however, bacterial resistance against antibiotics is rising even faster. For this reason, a complete shift of the antibacterial treatment paradigm is necessary. The development of technology has allowed us to rediscover well-known agents presenting antimicrobial properties with a better outcome. In this context, silver nanoparticles are a promising candidate for use in such therapy. Silver has many useful properties that can be used in the treatment of chronic wounds, such as anti-bacterial, anti-inflammatory, and anti-oxidative properties. In the form of nanoparticles, silver agents can work even more effectively and can be more easily incorporated into various dressings. Silver-based dressings are already commercially available; however, innovative combinations are still being discovered and very promising results have been described. In this review article, the authors focused on describing experimental and clinical studies exploring dressings containing either silver or silver nanoparticles, the results of which have been published in recent years.
Collapse
|
6
|
Assar DH, Mokhbatly AAA, Ghazy EW, Elbialy ZI, Gaber AA, Hassan AA, Nabil A, Asa SA. Silver nanoparticles induced hepatoxicity via the apoptotic/antiapoptotic pathway with activation of TGFβ-1 and α-SMA triggered liver fibrosis in Sprague Dawley rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:80448-80465. [PMID: 35716303 PMCID: PMC9596550 DOI: 10.1007/s11356-022-21388-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/06/2022] [Indexed: 06/15/2023]
Abstract
Despite the extraordinary use of silver nanoparticles (AgNPs) in medicinal purposes and the food industry, there is rising worry about potential hazards to human health and the environment. The existing study aims to assess the hepatotoxic effects of different dosages of AgNPs by evaluating hematobiochemical parameters, oxidative stress, liver morphological alterations, immunohistochemical staining, and gene expression to clarify the mechanism of AgNPs' hepatic toxic potential. Forty male Sprague Dawley rats were randomly assigned into control and three AgNPs intraperitoneally treated groups 0.25, 0.5, and 1 mg/kg b.w. daily for 15 and 30 days. AgNP exposure reduced body weight, caused haematological abnormalities, and enhanced hepatic oxidative and nitrosative stress with depletion of the hepatic GSH level. Serum hepatic injury biomarkers with pathological hepatic lesions where cholangiopathy emerges as the main hepatic alteration in a dosage- and duration-dependent manner were also elevated. Furthermore, immunohistochemical labelling of apoptotic markers demonstrated that Bcl-2 was significantly downregulated while caspase-3 was significantly upregulated. In conclusion, the hepatotoxic impact of AgNPs may be regulated by two mechanisms, implying the apoptotic/antiapoptotic pathway via raising BAX and inhibiting Bcl-2 expression levels in a dose-dependent manner. The TGF-β1 and α-SMA pathway which triggered fibrosis with incorporation of iNOS which consequently activates the inflammatory process were also elevated. To our knowledge, there has been no prior report on the experimental administration of AgNPs in three different dosages for short and long durations in rats with the assessment of Bcl-2, BAX, iNOS, TGF-β1, and α-SMA gene expressions.
Collapse
Affiliation(s)
- Doaa H. Assar
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Abd-Allah A. Mokhbatly
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Emad W. Ghazy
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Zizy I. Elbialy
- Department of Fish Processing and Biotechnology, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Ahmed A. Gaber
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Ayman A. Hassan
- High Technological Institute of Applied Health Sciences, Egypt Liver Research Institute and Hospital (ELRIAH), Sherbin, El Mansora Egypt
| | - Ahmed Nabil
- Beni-Suef University, Beni-Suef, Egypt
- Egypt Liver Research Institute and Hospital (ELRIAH), Sherbin, El Mansora Egypt
| | - Samah Abou Asa
- Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| |
Collapse
|
7
|
Cakir SN, Whitehead KM, Hendricks HKL, de Castro Brás LE. Novel Techniques Targeting Fibroblasts after Ischemic Heart Injury. Cells 2022; 11:cells11030402. [PMID: 35159212 PMCID: PMC8834471 DOI: 10.3390/cells11030402] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 12/12/2022] Open
Abstract
The great plasticity of cardiac fibroblasts allows them to respond quickly to myocardial injury and to contribute to the subsequent cardiac remodeling. Being the most abundant cell type (in numbers) in the heart, and a key participant in the several phases of tissue healing, the cardiac fibroblast is an excellent target for treating cardiac diseases. The development of cardiac fibroblast-specific approaches have, however, been difficult due to the lack of cellular specific markers. The development of genetic lineage tracing tools and Cre-recombinant transgenics has led to a huge acceleration in cardiac fibroblast research. Additionally, the use of novel targeted delivery approaches like nanoparticles and modified adenoviruses, has allowed researchers to define the developmental origin of cardiac fibroblasts, elucidate their differentiation pathways, and functional mechanisms in cardiac injury and disease. In this review, we will first characterize the roles of fibroblasts in the different stages of cardiac repair and then examine novel techniques targeting fibroblasts post-ischemic heart injury.
Collapse
Affiliation(s)
- Sirin N Cakir
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Kaitlin M Whitehead
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Hanifah K L Hendricks
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Lisandra E de Castro Brás
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
8
|
Kim S, Gates BL, Chang M, Pinkerton KE, Van Winkle L, Murphy CJ, Leonard BC, Demokritou P, Thomasy SM. Transcorneal delivery of topically applied silver nanoparticles does not delay epithelial wound healing. NANOIMPACT 2021; 24:100352. [PMID: 35559825 DOI: 10.1016/j.impact.2021.100352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 06/15/2023]
Abstract
Silver nanoparticles (AgNPs) are a common antimicrobial additive for a variety of applications, including wound care. However, AgNPs often undergo dissolution resulting in release of silver ions, with subsequent toxicity to mammalian cells. The cornea is a primary exposure site to topically administered AgNPs in and around the eye but their impact on corneal wound healing is understudied. Thus, the purpose of this study was to determine in vitro toxicity of AgNPs on corneal epithelial cells and fibroblasts as well as their effects on corneal epithelial wound healing utilizing an in vivo rabbit model. Non-coated 20 nm sized AgNP (AgNP-20) as well as 1% and 10% silver silica NPs (AgSiO2NPs) were tested at concentrations ranging from 0.05-250 μg/mL. Immortalized human corneal epithelial (hTCEpi) cells and primary rabbit corneal fibroblasts (RCFs) were incubated for 24 h with AgNPs and cell viability was tested. Additionally, a round wound healing assay was performed to determine hTCEpi cell migration. Quantitative real-time PCR and western blot analysis was performed to determine α-smooth muscle actin (α-SMA, a myofibroblast marker) mRNA and protein expression, respectively, in RCFs treated with 50 μg/mL of AgNPs. Corneal epithelial wound healing was evaluated with 1%-AgSiO2NPs (10 and 250 μg/mL) using an in vivo rabbit model. Rabbits were subsequently euthanized, and histologic sections of the enucleated globes were used to determine corneal penetration of 1%-AgSiO2NPs with autometallography and hyperspectral darkfield microscopy. Cell viability of both the hTCEpi cells and fibroblasts was significantly decreased by the three AgNPs in a dose dependent manner. Migration of hTCEpi cells was significantly inhibited by the three AgNPs. Alpha-SMA mRNA expression was significantly inhibited with three AgNPs, but only the 1%-AgSiO2NPs inhibited protein expression of α-SMA. In vivo epithelial wound closure did not significantly differ between groups treated with 10 or 250 μg/mL of 1%-AgSiO2NPs or vehicle control. The 1%-AgSiO2NPs penetrated throughout all corneal layers and into the anterior chamber in all treated eyes with no histopathological changes observed. In conclusion, the 1%-AgSiO2NPs are safe and have potential therapeutic applications through its efficacy of the corneal penetration and reduced scar formation during corneal wound healing.
Collapse
Affiliation(s)
- Soohyun Kim
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, Davis, CA 95616, USA
| | - Brooke L Gates
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, Davis, CA 95616, USA
| | - Maggie Chang
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, Davis, CA 95616, USA
| | - Kent E Pinkerton
- Center for Health and the Environment, University of California, Davis, CA 95616, USA
| | - Laura Van Winkle
- Center for Health and the Environment, University of California, Davis, CA 95616, USA; Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Christopher J Murphy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, Davis, CA 95616, USA; Department of Ophthalmology and Vision Science, School of Medicine, University of California, Davis, CA 95616, USA
| | - Brian C Leonard
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, Davis, CA 95616, USA
| | - Philip Demokritou
- Center for Nanotechnology and Nanotoxicology, HSPH-NIEHS Nanosafety Center, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, 665 Huntington, Boston, MA 02115, USA
| | - Sara M Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, Davis, CA 95616, USA; Department of Ophthalmology and Vision Science, School of Medicine, University of California, Davis, CA 95616, USA.
| |
Collapse
|
9
|
Rakowski M, Porębski S, Grzelak A. Silver Nanoparticles Modulate the Epithelial-to-Mesenchymal Transition in Estrogen-Dependent Breast Cancer Cells In Vitro. Int J Mol Sci 2021; 22:9203. [PMID: 34502112 PMCID: PMC8431224 DOI: 10.3390/ijms22179203] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/15/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
Silver nanoparticles (AgNPs) are frequently detected in many convenience goods, such as cosmetics, that are applied directly to the skin. AgNPs accumulated in cells can modulate a wide range of molecular pathways, causing direct changes in cells. The aim of this study is to assess the capability of AgNPs to modulate the metastasis of breast cancer cells through the induction of epithelial-to-mesenchymal transition (EMT). The effect of the AgNPs on MCF-7 cells was investigated via the sulforhodamine B method, the wound healing test, generation of reactive oxygen species (ROS), the standard cytofluorimetric method of measuring the cell cycle, and the expression of EMT marker proteins and the MTA3 protein via Western blot. To fulfill the results, calcium flux and HDAC activity were measured. Additionally, mitochondrial membrane potential was measured to assess the direct impact of AgNPs on mitochondria. The results indicated that the MCF-7 cells are resistant to the cytotoxic effect of AgNPs and have higher mobility than the control cells. Treatment with AgNPs induced a generation of ROS; however, it did not affect the cell cycle but modulated the expression of EMT marker proteins and the MTA3 protein. Mitochondrial membrane potential and calcium flux were not altered; however, the AgNPs did modulate the total HDAC activity. The presented data support our hypothesis that AgNPs modulate the metastasis of MCF-7 cells through the EMT pathway. These results suggest that AgNPs, by inducing reactive oxygen species generation, alter the metabolism of breast cancer cells and trigger several pathways related to metastasis.
Collapse
Affiliation(s)
- Michał Rakowski
- The Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, University of Lodz, 90-237 Lodz, Poland
- Cytometry Laboratory, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| | - Szymon Porębski
- Cytometry Laboratory, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| | - Agnieszka Grzelak
- Cytometry Laboratory, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| |
Collapse
|
10
|
Xie Y, Zhu S, Zang J, Wu G, Wen Y, Liang Y, Long Y, Guo W, Zang C, Hu X, Fan G, Xiang S, Zhang J. ADNP prompts the cisplatin-resistance of bladder cancer via TGF-β-mediated epithelial-mesenchymal transition (EMT) pathway. J Cancer 2021; 12:5114-5124. [PMID: 34335928 PMCID: PMC8317519 DOI: 10.7150/jca.58049] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/19/2021] [Indexed: 12/31/2022] Open
Abstract
Activity-dependent neuroprotective protein (ADNP) is vital for embryonic development and brain formation. Besides, the upregulated expression of ADNP enhances tumorigenesis in some human tumors like bladder cancer (BC). However, the potential roles of ADNP in drug resistance and the related mechanisms in BC is unknown. We performed this study to elucidate the influence of ADNP in the chemoresistance of BC and tried to explore the underlying molecular mechanism. The expressions of ADNP in BC from progression and non-progression patient specimens were measured by quantitative real-time PCR (qRT-PCR) and immunohistochemistry (IHC). In vitro experiments including colony formation, cell counting kit-8 (CCK-8), wound healing, and in vivo tumorigenesis assay were performed to explore the effects of ADNP on chemoresistance of BC. The impacts of ADNP on TGF-β/Smad signaling pathways were explored by western blot. Our results showed that the expression of ADNP mRNA and protein were significantly upregulated in BC tissues of the patients who suffered tumor-progression via RT-PCR and western blot. Cox regression survival analysis revealed that patients with high ADNP expression closely linked to shorter tumor-free survival. ADNP downregulation in BC showed more sensitive to cisplatin in vivo, while ADNP overexpression showed the opposite results. Additionally, we confirmed that ADNP promoted cell migration and EMT, thereby inducing cisplatin resistance, which may be related to TGF-β / Smad signaling pathway.
Collapse
Affiliation(s)
- Yu Xie
- Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, Hunan Normal University, 410081 Changsha, China.,Department of Urology, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, 410013 Changsha, China
| | - Shuai Zhu
- Department of Urology, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, 410013 Changsha, China
| | - Jinglei Zang
- Changsha Health Vocational College, 410600 Changsha, China
| | - Guanlin Wu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, 200433 Shanghai, China
| | - Yuheng Wen
- Department of Urology, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, 410013 Changsha, China
| | - Yu Liang
- Department of Urology, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, 410013 Changsha, China.,Pingxiang Maternal and Child Care Hospital, 337000 Pingxiang, China
| | - Ying Long
- Clinical Translational Research Center, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, 410013 Changsha, China
| | - Weiming Guo
- The 2nd Affiliated Hospital of South China University, 421001 Hengyang, China
| | - Chuanbing Zang
- Medizinische Klinik m. S. Hämatologie u. Onkologie, Campus Bejamin Franklin, Unviersitätsmedizin Berlin Charité, 12203 Berlin, Germany
| | - Xiang Hu
- Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, Hunan Normal University, 410081 Changsha, China
| | - Gang Fan
- Department of Urology, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, 410013 Changsha, China.,Department of Urology, Huazhong University of Science and Technology Union Shenzhen Hospital; the 6th Affiliated Hospital of Shenzhen University Health Science Center, 518060 Shenzhen, China
| | - Shuanglin Xiang
- Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, Hunan Normal University, 410081 Changsha, China
| | - Jian Zhang
- Key Laboratory of Protein Chemistry and Developmental Biology of the Ministry of Education, Hunan Normal University, 410081 Changsha, China
| |
Collapse
|
11
|
Yeyeodu ST, Martin ME, Reaves DK, Enders JR, Costantini LM, Fleming JM. Experimental data demonstrating the effects of silver nanoparticles on basement membrane gene and protein expression in cultured colon, mammary and bronchial epithelia. Data Brief 2019; 26:104464. [PMID: 31667234 PMCID: PMC6811978 DOI: 10.1016/j.dib.2019.104464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/18/2019] [Accepted: 08/23/2019] [Indexed: 12/01/2022] Open
Abstract
This data article is related to the research article entitled “Silver nanoparticles alter epithelial basement membrane integrity, cell adhesion molecule expression and TGF-beta secretion”, available in the journal Nanomedicine: Nanotechnology, Biology, and Medicine [1]. This Data in Brief consists of data that describe changes in the expression of basement membrane (BM)-associated genes and proteins in three non-transformed epithelial cell lines following acute (6 h) and chronic (24 h plus 7-day chase) exposure to silver nanoparticles (AgNPs). Human BEAS2B (lung), MCF10AI (breast), and CCD-18Co (colon) cultured epithelia were analyzed for protein expression by LC-MS/MS and for gene expression by pathway-focused QRT-PCR arrays of 168 focal adhesion, integrin, and extracellular matrix (ECM) genes known to be localized to the plasma membrane, the BM/ECM, or secreted into the extracellular space. Ingenuity pathway analysis (IPA) of combined gene and protein expression datasets was then used to predict canonical pathways affected by AgNP exposure.
Collapse
Affiliation(s)
| | - Megan E Martin
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC, USA
| | - Denise K Reaves
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC, USA
| | - Jeffrey R Enders
- Molecular Education, Technology and Research Innovation Center, North Carolina State University, Raleigh, NC, USA.,Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| | - Lindsey M Costantini
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC, USA
| | - Jodie M Fleming
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC, USA.,Molecular Education, Technology and Research Innovation Center, North Carolina State University, Raleigh, NC, USA.,Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|