Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Toxicol Appl Pharmacol ; 490: 117021, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38971382

ABSTRACT

Prostate cancer is a common type of cancer in men with high incidence and mortality. Our aim was to investigate the effects of oxalipalladium (ox-Pd) on metastatic human prostate cancer PC3 cells and compare them with the effects of oxaliplatin (ox-Pt) (as an approved cancer drug). We synthesized ox-Pd through a new chemical method and used FT-IR, 1H NMR, 13C NMR, and MS analyzes to characterize it. The effects of ox-Pd on PC3 cells viability, apoptosis, cell cycle, migration, and gene expression were examined. Inhibition of topoisomerase IIα activity was investigated by pHOT1 plasmid relaxation and kDNA decatenation assays. Chemical tests showed ox-Pd with the correct composition and structure. For the first time, the exact fragmentation pathway of ox-Pd and its difference with ox-Pt was obtained by MS analysis. Ox-Pd significantly decreased PC3 cell viability with less/no toxicity effect on MHFB-1 normal skin fibroblasts. Wound scratch assay confirmed the strong anti-migratory activity of ox-Pd. According to flow cytometry analysis, this drug increased the number of PC3 cells in late apoptosis and decreased DNA replication and mitosis. Furthermore, pHOT1 plasmid relaxation and kDNA decatenation assays showed that ox-Pd strongly inhibited the catalytic activity of topoisomerase IIα. The expression of topoisomerase IIα, Bcl-2, P21, and survivin was decreased while the expression of Bax and p53 was increased under ox-Pd treatment. We provide the first evidence that ox-Pd exhibits more selective anticancer effects on PC3 cells compared to ox-Pt. Taken together, these data strongly suggest a therapeutic window for ox-Pd in cancer.

2.
Fitoterapia ; 174: 105875, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38417678

ABSTRACT

Grapefruit mint (Mentha suaveolens × piperita) is a hybrid, perennial, and aromatic plant widely cultivated all over the world and used in the food, cosmetics, and pharmaceutical industries mostly for its valuable essential oil. Herein, we evaluated the anticancer activity of the grapefruit mint essential oil, cultivated in Iran. For the chemical composition analysis of essential oil, GC-MS was used. MTT assay was utilized for assessing the cytotoxic activity of the essential oil. The type of cell death was determined by annexin V/PI staining. Essential oil effect on the expression of maternally expressed gene 3 (MEG3), a regulatory lncRNA involved in cell growth, proliferation, and metastasis, was studied using qRT-PCR. Linalool (43.9%) and linalool acetate (40.1%) were identified as the dominant compounds of essential oil. Compared with MCF-7, the MDA-MB-231 cells were more sensitive to essential oil (IC50 = 7.6 µg/ml in MCF-7 and 5.9 µg/ml in MDA-MB-231 after 48 h). Essential oil induced cell death by apoptosis. Wound healing scratch assay confirmed the anti-invasive effect of essential oil. In addition, essential oil upregulated the tumor suppressor MEG3 in breast cancer cells. These results provide new insights into grapefruit mint essential oil potential application as an anticancer adjuvant in combination treatments for breast cancer patients.


Subject(s)
Acyclic Monoterpenes , Breast Neoplasms , Citrus paradisi , Mentha , Oils, Volatile , Humans , Female , Oils, Volatile/pharmacology , Oils, Volatile/chemistry , Mentha/chemistry , Molecular Structure , Breast Neoplasms/drug therapy , Mentha piperita
3.
Mol Biotechnol ; 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38349458

ABSTRACT

Vascular endothelial growth factor A165 (VEGF-A165) and VEGF receptor 2 (KDR) are important mediators of angiogenesis. We aimed to express the soluble KDR ligand-binding domain (sKDR1-3) and evaluate its interaction with the VEGF-A165 receptor-binding domain (VEGFA165-RBD). sKDR1-3 DNA was designed and subcloned into pPinkα-HC plasmid. The cassette was transfected into the Pichia pink™ 4 genome by homologous recombination. We optimized the expression of sKDR1-3 under the induction of different methanol concentrations. VEGFA165-RBD was expressed in E. coli BL21 harboring pET28a( +)─VEGFA165-RBD vector under induction with IPTG with/without lactose. Interaction and biological activity of sKDR1-3 and VEGFA165-RBD were investigated by ELISA and anti-proliferation tests. sKDR1-3 migrated on SDS-PAGE gel as a 35-180 kDa protein due to glycosylation. The relative expression level of sKDR1-3 under 1% methanol was higher than 0.5% and 4% methanol induction. IPTG and cysteine were suitable for induction and refolding of VEGFA165-RBD. 25 ng sKDR1-3 and 20 ng VEGFA165-RBD showed strong binding. sKDR1-3 bound to VEGFA165-RBD and VEGF-A165 with dissociation constants of 0.148 and 0.2 nM, respectively. 4-10 nM concentrations of sKDR1-3 inhibited the proliferation of HUVE cells induced by 5 nM VEGFA165-RBD. In consideration, sKDR1-3 in the nanomolar concentration range, is a promising anticancer drug to inhibit angiogenesis.

4.
Adv Pharm Bull ; 13(2): 385-392, 2023 Mar.
Article in English | MEDLINE | ID: mdl-37342383

ABSTRACT

Purpose: Non-viral transfection approaches are extensively used in cancer therapy. The future of cancer therapy lies on targeted and efficient drug/gene delivery. The aim of this study was to determine the transfection yields of two commercially available transfection reagents (i.e. Lipofectamine 2000, as a cationic lipid and PAMAM G5, as a cationic dendrimer) in two breast cell lines: cancerous cells (T47D) and non-cancerous ones (MCF-10A). Methods: We investigated the efficiencies of Lipofectamine 2000 and PAMAM G5 for transfection/delivery of a labeled short RNA into T47D and MCF-10A. In addition to microscopic assessments, the cellular uptakes of the complexes (fluorescein tagged-scrambled RNA with Lipofectamine or PAMAM dendrimer) were quantified by flow cytometry. Furthermore, the safety of the mentioned reagents was assessed by measuring cell necrosis through the cellular PI uptake. Results: Our results showed significantly better efficiencies of Lipofectamine compared to PAMAM dendrimer for short RNA transfection in both cell types. On the other hand, MCF-10A resisted more than T47D to the toxicity of higher concentrations of the transfection reagents. Conclusion: Altogether, our research demonstrated a route for comprehensive epigenetic modification of cancer cells and depicted an approach to efficient drug delivery, which eventually improves both short RNA-based biopharmaceutical industry and non-viral strategies in epigenetic therapy.

5.
Sci Rep ; 13(1): 9963, 2023 06 20.
Article in English | MEDLINE | ID: mdl-37339980

ABSTRACT

The skin undergoes the formation of fine lines and wrinkles through the aging process; also, burns, trauma, and other similar circumstances give rise to various forms of skin ulcers. Induced pluripotent stem cells (iPSCs) have become promising candidates for skin healing and rejuvenation due to not stimulating inflammatory responses, low probability of immune rejection, high metabolic activity, good large-scale production capacity and potentials for personalized medicine. iPSCs can secrete microvesicles (MVs) containing RNA and proteins responsible for the normal repairing process of the skin. This study aimed to evaluate the possibility, safety and effectiveness of applying iPSCs-derived MVs for skin tissue engineering and rejuvenation applications. The possibility was assessed using the evaluation of the mRNA content of iPSC-derived MVs and the behavior of fibroblasts after MV treatment. Investigating the effect of microvesicle on stemness potential of mesenchymal stem cells was performed for safety concerns. In vivo evaluation of MVs was done in order to investigate related immune response, re-epithelialization and blood vessel formation to measure effectiveness. Shedding MVs were round in shape distributed in the range from 100 to 1000 nm in diameter and positive for AQP3, COL2A, FGF2, ITGB, and SEPTIN4 mRNAs. After treating dermal fibroblasts with iPSC-derived MVs, the expressions of collagens Iα1 and III transcripts (as the main fibrous extracellular matrix (ECM) proteins) were upregulated. Meanwhile, the survival and proliferation of MV treated fibroblasts did not change significantly. Evaluation of stemness markers in MV treated MSCs showed negligible alteration. In line with in vitro results, histomorphometry and histopathology findings also confirmed the helpful effect of MVs in skin regeneration in the rat burn wound models. Conducting more investigations on hiPSCs-derived MVs may lead to produce more efficient and safer biopharmaceutics for skin regeneration in the pharmaceutical market.


Subject(s)
Cell-Derived Microparticles , Induced Pluripotent Stem Cells , Humans , Rats , Animals , Induced Pluripotent Stem Cells/metabolism , Transcriptome , Rejuvenation , Skin/pathology , Cell-Derived Microparticles/metabolism
7.
J Biomater Appl ; 37(8): 1341-1354, 2023 03.
Article in English | MEDLINE | ID: mdl-36331116

ABSTRACT

The developing fetus is wrapped by a human amniotic membrane or amnion. Amnion is a promising human tissue allograft in clinical application because of its chemical composition, collagen-based, and mechanical properties of the extracellular matrix. In addition, amnion contains cells and growth factors; therefore, meets the essential parameters of tissue engineering. No donor morbidity, easy processing and storage, fewer ethical issue, anti-inflammatory, antioxidant, antibacterial, and non-immunogenic properties are other advantages of amnion usage. For these reasons, amnion can resolve some bottlenecks in the regenerative medicine issues such as tissue engineering and cell therapy. Over the last decades, biomedical applications of amnion have evolved from a simple sheet for skin or cornea repair to high-technology applications such as amnion nanocomposite, powder, or hydrogel for the regeneration of cartilage, muscle, tendon, and heart. Furthermore, amnion has anticancer as well as drug/cell delivery capacity. This review highlights various ancient and new applications of amnion in research and clinical applications, from regenerative medicine to cancer therapy, focusing on articles published during the last decade that also revealed information regarding amnion-based products. Challenges and future perspectives of the amnion in regenerative medicine are also discussed.


Subject(s)
Amnion , Regenerative Medicine , Humans , Amnion/chemistry , Tissue Engineering , Skin
8.
Appl Microbiol Biotechnol ; 106(17): 5335-5347, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35871694

ABSTRACT

Metabolic differences between normal and cancerous cells have been used as a point of view for developing anticancer drugs. Some degrading enzymes of certain amino acids have been regarded to kill cancerous cells. L-Asparaginase (ASNase) has shown an excellent therapeutic response to asparagine-auxotrophic cancers such as acute lymphoblastic leukemia (ALL). Some bacteria, yeasts, molds, plants, and animals produce ASNase. Bacterial ASNases from Escherichia coli and Erwinia chrysanthemi are the FDA-approved drugs for ALL treatment. Here, we review new natural prokaryotic and eukaryotic sources of ASNases, recent advances to introduce improvement strategies for the production of recombinant ASNases as well as their chemical modifications, immobilization, nanoencapsulation, and in silico studies to increase efficiency and decrease side effects. Recent studies for application of ASNases to treatment of asparagine-auxotrophic cancers, especially solid cancers, have been reviewed. Furthermore, challenges and future perspectives are discussed for this promising therapeutic enzyme. KEY POINTS: • Review recent advances to introduce new sources of microbial L-asparaginases. • Review improvement strategies for the development of stable and non-toxic L-asparaginases. • Review microbial L-asparaginase application in various cancers' treatment.


Subject(s)
Antineoplastic Agents , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Animals , Asparaginase , Asparagine , Bacteria , Escherichia coli
9.
Gene ; 808: 145963, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34530086

ABSTRACT

As of July 2021, the outbreak of coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2, has led to more than 200 million infections and more than 4.2 million deaths globally. Complications of severe COVID-19 include acute kidney injury, liver dysfunction, cardiomyopathy, and coagulation dysfunction. Thus, there is an urgent need to identify proteins and genetic factors associated with COVID-19 susceptibility and outcome. We comprehensively reviewed recent findings of host-SARS-CoV-2 interactome analyses. To identify genetic variants associated with COVID-19, we focused on the findings from genome and transcriptome wide association studies (GWAS and TWAS) and bioinformatics analysis. We described established human proteins including ACE2, TMPRSS2, 40S ribosomal subunit, ApoA1, TOM70, HLA-A, and PALS1 interacting with SARS-CoV-2 based on cryo-electron microscopy results. Furthermore, we described approximately 1000 human proteins showing evidence of interaction with SARS-CoV-2 and highlighted host cellular processes such as innate immune pathways affected by infection. We summarized the evidence on more than 20 identified candidate genes in COVID-19 severity. Predicted deleterious and disruptive genetic variants with possible effects on COVID-19 infectivity have been also summarized. These findings provide novel insights into SARS-CoV-2 biology and infection as well as potential strategies for development of novel COVID therapeutic targets and drug repurposing.


Subject(s)
COVID-19/metabolism , Host Microbial Interactions/genetics , SARS-CoV-2/metabolism , COVID-19/physiopathology , Computational Biology/methods , Cryoelectron Microscopy/methods , Crystallography, X-Ray/methods , Genome-Wide Association Study , Host Microbial Interactions/physiology , Host-Pathogen Interactions/genetics , Humans , Proteins/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity
10.
Curr Med Chem ; 29(23): 4078-4096, 2022.
Article in English | MEDLINE | ID: mdl-34825864

ABSTRACT

Cancer, a challenging medical problem, affects millions of people around the world. Cancer cell resistance is one of the main drawbacks in the complete prosperity of even more sophisticated therapies. Pore-forming peptides (PFPs), a group of natural defense system proteins are used by nearly all living organisms as anti-bacterial and anti-- fungal agents, and could also be regarded as novel tumoricidal peptides. PFPs approach entails using soluble peptides by assembling them mainly on the target cell membrane and forming potential death-causing pores. Physical damage induction by natural PFPs or their synthetic derivatives could conquer the resistance mechanisms of tumor cells. Given that peptide drugs involve a significant proportion of the pharmaceutical market primarily because of easy synthesis and safety, evaluating this nature provided a model system as a group of anticancer peptides seems a valuable approach. Here, the mode of action of PFPs and their anticancer mechanism are highlighted, followed by addressing the anticancer studies using PFPs from different sources along with various strategies applied to obtain selective action of PFPs against cancer cells. Challenges and future perspectives of these promising bioactive molecules in cancer treatment are also provided.


Subject(s)
Neoplasms , Peptides , Cell Membrane/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Peptides/analysis , Peptides/pharmacology , Peptides/therapeutic use
11.
Microb Pathog ; 154: 104831, 2021 May.
Article in English | MEDLINE | ID: mdl-33727169

ABSTRACT

The third pandemic of coronavirus infection, called COVID-19 disease, began recently in China. The newly discovered coronavirus, entitled SARS-CoV-2, is the seventh member of the human coronaviruses. The main pathogenesis of SARS-CoV-2 infection is severe pneumonia, RNAaemia, accompanied by glass turbidity, and acute cardiac injury. It possesses a single-stranded positive-sense RNA genome which is 60-140 nm in diameter, and has a size of 26-32 kbp. Viral pathogenesis is accomplished with spike glycoprotein through the employment of a membrane-bound aminopeptidase, called the ACE2, as its primary cell receptor. It has been confirmed that various factors such as different national rules for quarantine and various races or genetic backgrounds might influence the mortality and infection rate of COVID-19 in the geographic areas. In addition to various known and unknown factors and host genetic susceptibility, mutations and genetic variabilities of the virus itself have a critical impact on variable clinical features of COVID-19. Although the SARS-CoV-2 genome is more stable than SARS-CoV or MERS-CoV, it has a relatively high dynamic mutation rate with respect to other RNA viruses. It's noteworthy that, some mutations can be founder mutations and show specific geographic patterns. Undoubtedly, these mutations can drive viral genetic variability, and because of genotype-phenotype correlation, resulting in a virus with more/lower/no decrease in natural pathogenic fitness or on the other scenario, facilitating their rapid antigenic shifting to escape the host immunity and also inventing a drug resistance virus, so converting it to a more infectious or deadly virus. Overall, the detection of all mutations in SARS-CoV-2 and their relations with pathological changes is nearly impossible, mostly due to asymptomatic subjects. In this review paper, the reported mutations of the SARS-CoV-2 and related variations in virus structure and pathogenicity in different geographic areas and genotypes are widely investigated. Many studies need to be repeated in other regions/locations for other people to confirm the findings. Such studies could benefit patient-specific therapy, according to genotyping patterns of SARS-CoV-2 distribution.


Subject(s)
COVID-19 , SARS-CoV-2 , China/epidemiology , Humans , Mutation , Spike Glycoprotein, Coronavirus/genetics , Virulence
12.
Infect Genet Evol ; 90: 104773, 2021 06.
Article in English | MEDLINE | ID: mdl-33607284

ABSTRACT

The third pandemic of coronavirus infection, called COVID-19 disease, was first detected in November 2019th. Various determinants of disease progression such as age, sex, virus mutations, comorbidity, lifestyle, host immune response, and genetic background variation have caused clinical variability of COVID-19. The causative agent of COVID-19 is an enveloped coronavirus named severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) that invades host cells using an endocytic pathway. The SARS-CoV-2 spike protein is the main viral protein that contributes to the fusion of the virus particle to the host cell through angiotensin-converting enzyme 2 (ACE2). The highly conserved expression of ACE2 is found in various animals, which indicates its pivotal physiological function. The ACE2 has a crucial role in vascular, renal, and myocardial physiology. Genetic factors contributing to the outcome of SARS-CoV-2 infection are unknown; however, variants in the specific sites of ACE2 gene could be regarded as a main genetic risk factor for COVID-19. Given that ACE2 is the main site for virus landing on host cells, the effect of amino acid sequences of ACE2 on host susceptibility to COVID-19 seems reasonable. It would likely have a substantial role in the occurrence of a wide range of clinical symptoms. Several ACE2 variants can affect the protein stability, influencing the interaction between spike protein and ACE2 through imposing conformational changes while some other variants are known to cause a decrease or an increase in the ligand-receptor affinity. The other variations are located at the proteolytic cleavage site, which can influence virus infection; because soluble ACE2 can act as a decoy receptor for virus and decrease virus intake by cell surface ACE2. Notably, polymorphisms of regulatory and non-coding regions such as promoter in ACE2, can play crucial role in different expression levels of ACE2 among different individuals. Many studies should be performed to investigate the involvement of ACE2 polymorphism with susceptibility to COVID-19. Herein, we discuss some reported associations between variants of ACE2 and COVID-19 in details. In addition, the mode of action of ACE2 and its role in SARS-CoV-2 infection are highlighted which is followed by addressing the effects of several ACE2 variants on its protein stability, viral tropism or ligand-receptor affinity, secondary and tertiary structure or protein conformation, proteolytic cleavage site, and finally inter-individual clinical variability in COVID-19. The polymorphisms of regulatory regions of ACE2 and their effect on expression levels of ACE2 are also provided in this review. Such studies can improve the prediction of the affinity of mutant ACE2 variations with spike protein, and help the biopharmaceutical industry to design effective approaches for recombinant hACE2 therapy and vaccination of COVID-19 disease.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , COVID-19/genetics , COVID-19/virology , Disease Susceptibility , Genetic Variation , Host-Pathogen Interactions , SARS-CoV-2/physiology , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/diagnosis , COVID-19/metabolism , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/metabolism , Disease Management , Host-Pathogen Interactions/immunology , Humans , Immune Evasion , Immunity, Innate , Polymorphism, Single Nucleotide , Prognosis , Protein Binding , Receptors, Virus/metabolism , Severity of Illness Index
13.
J Cell Physiol ; 235(5): 4153-4166, 2020 05.
Article in English | MEDLINE | ID: mdl-31647128

ABSTRACT

Despite many advances and optimization in colon cancer treatment, tumor recurrence and metastases make the development of new therapies necessary. Colon cancer stem cells (CCSCs) are considered as the main triggering factor of cancer progression, recurrence, and metastasis. CCSCs as a result of accumulated genetic and epigenetic alterations and also complex interconnection with the tumor microenvironment (TME) can evolve and convert to full malignant cells. Mounting evidence suggests that in cancer therapy both CCSCs and non-CCSCs in TME have to be regarded to break through the limitation of current therapies. In this regard, stem cell capabilities of some non-CCSCs may arise inside the TME condition. Therefore, a deep knowledge of regulatory mechanisms, heterogeneity, specific markers, and signaling pathways of CCSCs and their interconnection with TME components is needed to improve the treatment of colorectal cancer and the patient's life quality. In this review, we address current different targeted therapeutic options that target cell surface markers and signaling pathways of CCSCs and other components of TME. Current challenges and future perspectives of colon cancer personalized therapy are also provided here. Taken together, based on the deep understanding of biology of CCSCs and using three-dimensional culture technologies, it can be possible to reach successful colon cancer eradication and improvise combination targeted therapies against CCSCs and TME.


Subject(s)
Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Neoplastic Stem Cells/physiology , Tumor Microenvironment , Biomarkers, Tumor , Gene Expression Regulation, Neoplastic , Humans , Signal Transduction
14.
Drug Discov Today ; 25(3): 552-560, 2020 03.
Article in English | MEDLINE | ID: mdl-31843577

ABSTRACT

Nucleic acid vaccines (NAVs) have recently been tested as a cancer therapy. DNA and mRNA vaccines deliver genetic information encoding tumor antigens (TAs) to the host, which then produces immune responses against cancer cells that express the TAs. Although NAVs are easy, safe, and simple to manufacture, they have not so far been considered viable alternatives to peptide vaccines. Choosing the right TAs, insufficient immunogenicity, and the immunosuppressive nature of cancer are some challenges to this approach. In this review, we discuss approaches that been used to improve the efficiency of anticancer NAVs.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Neoplasms/therapy , Animals , Cancer Vaccines/immunology , Humans , Immunogenicity, Vaccine , Neoplasms/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , mRNA Vaccines
15.
J Cell Biochem ; 120(10): 17194-17207, 2019 10.
Article in English | MEDLINE | ID: mdl-31104319

ABSTRACT

BACKGROUND: Loss of skin integrity due to injury, burning, or illness makes the development of new treatment options necessary. Skin tissue engineering provides some solutions for these problems. OBJECTIVE: The potential of a biodegradable star-shaped copolymer [Poly(CL─CO─LA)-b-PEG] and penta-block copolymer hydrogel (PNIPAAm-PCL-PEG-PCL-PNIPAAm) was assessed for skin tissue engineering applications. METHODS: Two copolymers were synthesized for cellular culture scaffolds and their mechanical properties were compared. The resulting star-shaped copolymer and thermosensitive penta-block copolymer were characterized using Fourier transform infrared and nuclear magnetic resonance spectroscopy. The crystallizability of the two copolymers was analyzed using X-ray diffraction. The resulting thermosensitive penta-block copolymer was evaluated by differential thermal analysis, differential scanning calorimetry and thermogravimetric analysis. Scanning electron microscopy and in vitro degradation of the polymer network in phosphate buffer solutions (pH 7.4) at 37°C were also examined. The pore size of the gels was calculated with Image Analyzer software. Finally, the cytotoxic, morphological, and gene expression effects of copolymers on the skin fibroblast were evaluated. RESULTS: The experiments showed that the PNIPAAm-PCL-PEG-PCL-PNIPAAm polymer with the right composition and the expected molecular weight was achieved. The hydrogel had less crystallizability compared with its precursors. The resulting thermosensitive hydrogel had a three-dimensional structure with interconnected pores that mimicked the extracellular matrix. The control of the degradability rate can be possible by weight percent changes. The pore size correlated with the polymer concentration in aqueous solution and the pore sizes of the 20 wt% hydrogel were better for fibroblast cultivation than those of the 10 wt% hydrogel. Cell proliferation on the 20% gel was more than that of the 10% gel. The hydrogel not only preserved the viability and phenotypical morphology of the entrapped cells but also stimulated the initial cell-cell interactions and proliferation of fibroblasts. The hydrogel did not influence cell conformation and this property of the polymer underlined its safety. Cells seeded on this copolymer showed a normal and spear shape and formed a focal adhesion with the hydrogel surface. Notably, the hydrogel increased collagen I α1 and collagen III mRNAs expression. CONCLUSION: Due to the low molecular weight and poor mechanical strength of the star-shaped copolymer, it was not considered for fabrication of the scaffolds for wound healing. The biodegradable, biocompatible, injectable and thermosensitive PNIPAAm-PCL-PEG-PCL-PNIPAAm hydrogel in 20 wt% demonstrated a desirable potential for future application as a cell scaffold in skin tissue engineering and wound healing.


Subject(s)
Absorbable Implants , Fibroblasts/drug effects , Hydrogels/chemical synthesis , Polyesters/chemical synthesis , Polyethylene Glycols/chemical synthesis , Acrylic Resins/chemistry , Biomarkers/metabolism , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Collagen Type I/agonists , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Collagen Type III/agonists , Collagen Type III/genetics , Collagen Type III/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression/drug effects , Humans , Hydrogels/pharmacology , Polyesters/pharmacology , Polyethylene Glycols/pharmacology , Porosity , RNA, Messenger/agonists , RNA, Messenger/genetics , RNA, Messenger/metabolism , Skin/cytology , Temperature , Tissue Engineering/methods , Wound Healing/drug effects
16.
Life Sci ; 213: 236-247, 2018 Nov 15.
Article in English | MEDLINE | ID: mdl-30308184

ABSTRACT

Silibinin is a natural plant polyphenol with high antioxidant and anticancer properties, which causes broad-spectrum efficacy against cancer, including cell cycle arrest and apoptosis in most cancer cell types. Silibinin, by modulating the apoptosis, cell cycle progression and autophagic pathways in various cellular and molecular routs might be used to design more effective anticancer strategies. Silibinin also regulates aberrant miRNAs expression linked to many aspects of cell biology in cancer. Maybe the most interesting aspect of silibinin is its ability to trigger multiple cellular signaling pathways to induce a particular biologic effect in various cell types. This review discusses investigations supporting the ability of silibinin to be as a natural modulator of involved cellular biological events in cancer progression. In this review, we introduce the salient features of silibinin therapy to optimize clinical outcomes for oncology patients. The goal of the treatments is to make it possible to eliminate the tumor with the minimum side effects and cure the patient in the early stage cancer. Therefore, plant extracts such as silibinin can be included in the treatments.


Subject(s)
Neoplasms/drug therapy , Silybin/metabolism , Silybin/pharmacology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Division/drug effects , Cell Line, Tumor , Flavonolignans/pharmacology , Humans , MicroRNAs/drug effects , Signal Transduction/drug effects
17.
Phytomedicine ; 49: 32-40, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-30217260

ABSTRACT

BACKGROUND: Chordoma, slow growing bone tumours originating from remnants of the notochord, leave affected patients with a median survival of six years. The high recurrence rate of chordoma, together with limited treatment options and bad overall prognosis, make the development of new treatment options urgently necessary. PURPOSE: In this study, the potential of two natural products, silibinin and ß-ß-dimethylacrylshikonin (DMAS), was tested on clival (MUG-CC1 and UM-Chor1) as well as sacral (MUG-Chor1 and U-CH2) chordoma cell lines. The treatment was administered both as single- and combined therapy. METHODS: For investigation of cell viability, the Cell Titer 96 Aqueous Non-Radioactive Cell Proliferation Assay Kit was used. Apoptosis induction was studied by flow cytometry, (Annexin V/SYTOX Green, caspase-3) and RT-qPCR. Pathway analyses were performed by western blot. RESULTS: Both drugs were found to reduce cell viability alone as well as in combination in a dose dependent manner, with DMAS being more efficient than silibinin. The mode of cell death was mainly apoptosis in DMAS treated samples, while the combination therapy led to apoptosis as well as late-apoptosis/necrosis. Silibinin therapy alone, although reducing cell viability, did not lead to significant apoptotic effects in the performed assays. Focussing on the molecular mechanism of DMAS induced apoptosis, it was found that major genes of the mitochondrial apoptosis pathway, like NOXA and PUMA were overexpressed. Additionally, western blot experiments showed a decrease of ERK/pERK, STAT3/pSTAT3 (Tyr705) and AKT/pAKT expression/activation levels under DMAS treatment. CONCLUSION: DMAS is a promising new candidate for chordoma therapy, while silibinin or a combination of both is less favourable.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Bone Neoplasms/pathology , Chordoma/pathology , Naphthoquinones/pharmacology , Silymarin/pharmacology , Apoptosis/drug effects , Bone Neoplasms/drug therapy , Boraginaceae/chemistry , Caspase 3 , Cell Line, Tumor , Cell Survival/drug effects , Chordoma/drug therapy , Humans , Mitochondria/drug effects , Plant Roots/chemistry , Signal Transduction , Silybin
18.
Cytotechnology ; 69(4): 667-680, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28321778

ABSTRACT

Silibinin is a natural polyphenol with high antioxidant and anticancer properties, which causes cell cycle arrest and apoptosis in most cancer cell types including breast cancer, but the in-line mechanisms, are still unknown. Silibinin significantly downregulated oncomiR miR-21 expression in breast cancer cells. Here the effect of anti-miR-21 on cell viability, apoptotic induction, cell cycle distribution, and the expression levels of downstream targets of miR-21 were investigated in MCF-7 and T47D cells. MiR-21 mimic transfection was also applied in silibinin treated samples to evaluate functional role of miR-21downregulation on silibinin effects. It was found that after anti-miR-21 transfection, no significant changes were detected in cell viability, apoptosis (except early apoptosis), and cell cycle in MCF-7 and T47D cells. Compared to silibinin, miR-21 mimic transfection in combination with silibinin caused a slight modulation in some of the examined silibinin effects including apoptosis, Bcl2 mRNA and PTEN mRNA and protein levels. Silibinin slightly changed luciferase activity from reporters containing the miR-21 recognition elements from PTEN-3'UTR and Bcl2-3'UTR in both cell lines. Together these data demonstrated negligible cancer-progression impact of miR-21 and limited roles of miR-21 downregulation in examined silibinin effects, and strengthened the anti-cancer pathways of silibinin other than miR-21downregulation in MCF-7 and T47D cells.

19.
Asian Pac J Cancer Prev ; 17(5): 2661-5, 2016.
Article in English | MEDLINE | ID: mdl-27268647

ABSTRACT

Silibinin is a natural polyphenol with high antioxidant and anticancer properties. In this study, its influence on two of the most commonly employed human breast cancer cell lines, MCF-7 and T47D, and one non-malignant MCF-10A cell line, were investigated and compared. Cell viability, the cell cycle distribution and apoptosis induction were analyzed by MTT and flow cytometry, respectively. The effect of silibinin on PTEN, Bcl-2, P21, and P27 mRNAs expression was also investigated by real-time RT-PCR. It was found that silibinin caused G1 cell cycle arrest in MCF-7 and MCF-10A cells but had no effect on the T47D cell cycle. Silibinin induced cytotoxic and apoptotic effects in T47D cells more than the MCF-7 cells and had no cytotoxic effect in MCF-10A cells under the same conditions. Silibinin upregulated PTEN in MCF-7 and caused slightly increased P21 mRNA expression in T47D cells and slightly increased PTEN and P21 expression in MCF-10A cells. Bcl-2 expression decreased in all of the examined cells under silibinin treatment. P27 mRNA expression upregulated in T47D and MCF-10A cells under silibinin treatment. PTEN mRNA in T47D and P21 and P27 mRNAsin MCF-7 were not affected by silibinin. These results suggest that silibinin has mostly different inhibitory effects in breast cancer cells and might be an effective anticancer agent for some cells linked to influence on cell cycle progression.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Cycle/drug effects , Signal Transduction/drug effects , Silymarin/pharmacology , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Female , Humans , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Silybin , Tumor Cells, Cultured
20.
Afr J Paediatr Surg ; 11(3): 233-7, 2014.
Article in English | MEDLINE | ID: mdl-25047315

ABSTRACT

BACKGROUND: Biliary atresia (BA) is a progressive inflammatory destructive process of the bile ducts. This study evaluated the relationship between single-nucleotide polymorphisms in the promoter region of tumour necrosis factor-alpha (TNF-α) gene and bilaiary atresia. MATERIALS AND METHODS: Genomic deoxyribonucleic acid from 16 patients with established diagnosis of BA and 36 patients with INC was obtained. The genotypes of TNF-α-1031 (T/C) and TNF-α-308 (G/A) were determined using the restriction fragment length polymorphism-polymerase chain reaction and the results were analysis with proper statistic software. RESULTS: The frequencies of T/T, T/C in TNF-α-1031 and G/G, G/A in TNF-α-308 were as same as control group. Moreover, we have same deduction for allele frequency and haplotypes analysis (T allele: 84.37%; G allele: 87.5%) in BA patients (T allele: 80.56%; G allele: 86.11%) in controls. In all cases variants of polymorphism did not affect the severity or incidence of BA disease. CONCLUSION: although no significant associations were found between BA and control groups, it seems meaningful that since the nature of BA is multi factorial. Next step will be considering a new target such as downstream modulation of the TNF-α pathway or other cytokines and chemokines which act directly/indirectly.


Subject(s)
Biliary Atresia/genetics , DNA/genetics , Genetic Predisposition to Disease , Polymorphism, Genetic , Tumor Necrosis Factor-alpha/genetics , Alleles , Biliary Atresia/epidemiology , Female , Gene Frequency , Genotype , Humans , Incidence , Infant , Infant, Newborn , Iran/epidemiology , Male , Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...