Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Carbohydr Polym ; 326: 121614, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38142075

ABSTRACT

Massive bleeding control plays the main role in saving people's lives in emergency situations. Herein, modified cellulose-based nanocomposite sponges by polydopamine (PDA) and laponite nano-clay was developed to sturdily deal with non-compressible lethal severe bleeding. PDA accomplishes supreme adhesion in the bleeding site (∼405 kPa) to form strong physical barrier and seal the position. Sponges super porous (∼70 % porosity) and super absorbent capacity (48 g blood absorbed per 1 g sponge) by concentrating the blood cells and platelets provides the requirements for primary hemostasis. Synergistically, the nanocomposite sponges' intelligent chemical structure induces hemostasis by activation of the XI, IX, X, II and FVII factors of intrinsic and extrinsic coagulation pathways. Excellent hemostatic performance of sponges in-vitro was assessed by RBC accumulation (∼100 %), blood clotting index (∼10 %), platelet aggregation/activation (∼93 %) and clotting time. The nanocomposite sponges depicted super performance in the fatal high-pressure non-compressible hemorrhage model by reducing of >2, 15 and 3 times in the bleeding amount at New Zealand rabbit's heart and liver, and rat's femoral artery bleeding models, respectively compared to commercial hemostatic agents (Pvalue˂0.001). The in-vivo host response results exhibited biosafety with no systemic and significant local inflammatory response by hematological, pathological and biochemical parameters assessments.


Subject(s)
Hemostatics , Nanocomposites , Humans , Rabbits , Rats , Animals , Adhesives/pharmacology , Clay , Citric Acid , Hemostasis , Hemostatics/chemistry , Hemorrhage/drug therapy , Cellulose/pharmacology , Cellulose/chemistry , Nanocomposites/chemistry
2.
Sci Rep ; 12(1): 8316, 2022 05 18.
Article in English | MEDLINE | ID: mdl-35585092

ABSTRACT

As a platinum-containing anticancer drug, cisplatin is the keystone for treating many malignancies. Nephrotoxicity is the main dose-limiting toxicity, and several hydration therapies and supplementary strategies are utilized to reduce cisplatin-induced kidney damage, so the discovery and development of effective and safe antitumor drugs are still on the path of human health. Herein, a new four-coordinated Pt complex [Pt(TSC)Cl] using N(4)-phenyl-2-formylpyridine thiosemicarbazone (HTSC) was synthesized and characterized by single-crystal X-ray diffraction, 1HNMR, FT-IR, LC/MS and CHN elemental analysis. The Pt(TSC)Cl complex revealed antiproliferative activity against A549, MCF-7 and Caco-2 cell lines with a low micromolar IC50 (200-1.75 µM). Specifically, the Pt(TSC)Cl complex displayed more selectivity in Caco-2 cells (IC50 = 2.3 µM) than cisplatin (IC50 = 107 µM) after 48 h of treatment. Moreover, compared with cisplatin, a known nephrotoxic drug, the Pt(TSC)Cl complex exhibited lower nephrotoxicity against Hek293 normal cells. We also found that the Pt(TSC)Cl complex can effectively prevent cancer cell propagation in sub-G1 and S phases and induce apoptosis (more than 90%). Real time PCR and western analysis demonstrated that the expression pattern of apoptotic genes and proteins is according to the intrinsic apoptosis pathway through the Bax/Bcl-2-Casp9-Casp3/Casp7 axis. Collectively, our findings indicated that the Pt(TSC)Cl complex triggers apoptosis in Caco-2 cell lines, while low nephrotoxicity was shown and may be considered a useful anticancer drug candidate for colorectal cancers for further optimization and growth.


Subject(s)
Antineoplastic Agents , Cisplatin , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Apoptosis , Caco-2 Cells , Cell Line, Tumor , Cisplatin/adverse effects , HEK293 Cells , Humans , Spectroscopy, Fourier Transform Infrared
3.
Pathol Res Pract ; 216(12): 153247, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33190014

ABSTRACT

The most prevalent malignancy that can occur in the gastrointestinal tract is colon cancer. The current treatment options for colon cancer patients include chemotherapy, surgery, radiotherapy, immunotherapy, and targeted therapy. Although the chance of curing the disease in the early stages is high, there is no cure for almost all patients with advanced and metastatic disease. It has been found that over-activation of cyclooxygenase 2 (COX-2), followed by the production of prostaglandin E2 (PGE2) in patients with colon cancer are significantly increased. The tumorigenic function of COX-2 is mainly due to its role in the production of PGE2. PGE2, as a main generated prostanoid, has an essential role in growth and survival of colon cancer cell's. PGE2 exerts various effects in colon cancer cells including enhanced expansion, angiogenesis, survival, invasion, and migration. The signaling of PGE2 via the EP4 receptor has been shown to induce colon tumorigenesis. Moreover, the expression levels of the EP4 receptor significantly affect tumor growth and development. Overexpression of EP4 by various mechanisms increases survival and tumor vasculature in colon cancer cells. It seems that the pathway starting with COX2, continuing with PGE2, and ending with EP4 can promote the spread and growth of colon cancer. Therefore, targeting the COX-2/PGE2/EP4 axis can be considered as a worthy therapeutic approach to treat colon cancer. In this review, we have examined the role and different mechanisms that the EP4 receptor is involved in the development of colon cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Colonic Neoplasms/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Animals , Antineoplastic Agents/therapeutic use , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Humans , Ligands , Molecular Targeted Therapy , Prostaglandin Antagonists/therapeutic use , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Signal Transduction
4.
Adv Pharm Bull ; 10(4): 502-511, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33062601

ABSTRACT

Proprotein convertase subtilisin/kexin type 9 (PCSK9), as a vital modulator of low-density lipoprotein cholesterol (LDL-C) , is raised in hepatocytes and released into plasma where it binds to LDL receptors (LDLR), leading to their cleavage. PCSK9 adheres to the epidermal growth factor-like repeat A (EGF-A) domain of the LDLR which is confirmed by crystallography. LDLR expression is adjusted at the transcriptional level through sterol regulatory element binding protein 2 (SREBP-2) and at the post translational stages, specifically through PCSK9, and the inducible degrader of the LDLR PCSK9 inhibition is an appealing new method for reducing the concentration of LDL-C. In this review the role of PCSK9 in lipid homeostasis was elucidated, the effect of PCSK9 on atherosclerosis was highlighted, and contemporary therapeutic techniques that focused on PCSK9 were summarized. Several restoration methods to inhibit PCSK9 have been proposed which concentrate on both extracellular and intracellular PCSK9, and they include blockage of PCSK9 production by using gene silencing agents and blockage of it's binding to LDLR through antibodies and inhibition of PCSK9 autocatalytic processes by tiny molecule inhibitors.

5.
Colloids Surf B Biointerfaces ; 194: 111225, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32622253

ABSTRACT

This study aimed to design an effective targeted combination of doxorubicin (Dox)-Curcumin (Cur) delivery system to eradicate the MDA-MB231 cell line. A novel biodegradable poly ε-Caprolactone-co-maleic anhydride-graft-citric acid copolymer micelle (PCL-co-P(MA-g-CA)) was synthesized through thiolen radical copolymerization and ring-opening polymerization. The unique micelle structure allowed simultaneous loading of hydrophilic Dox and hydrophobic Cur with a loading efficiency of above 98 % for each drug. The physicochemical characterization of copolymeric micelle was analyzed by 1HNMR, 13CNMR, FTIR, DSC, CMC, DLS and SEM. The in vitro cytotoxicity was assessed by MTT assay, cell cycle analysis, annexin V-FITC apoptosis, qRT-PCR and western blot. The final obtained micelles with critical micelle concentration (CMC) of 0.5 µg/mL, and particle size and surface charge was 60 nm and -14.1 mV, respectively. Beside the fast uptake of designed micelle, Dox@Cur loaded micelle showed a synergistic effect with the combination index (CI) value of below 1. Our results revealed that this novel engineered combinatorial micelle induced apoptosis (96 %) which was proved by annexin V and cell cycle. qRT-PCR and western blot assays demonstrated involvement of intrinsic apoptosis pathways in the genetic and protein levels. Finally, the penetration of Dox@Cur loaded micelle was evaluated by 3D in vitro tumor formation. Our findings showed the penetration behavior of micelles is in a concentration-dependent manner. In conclusion, combinational therapy by using Dox and Cur nano-formulation has boosted the cytotoxicity in MDA-MB231 cells by promoting the apoptotic response.


Subject(s)
Curcumin , Micelles , Caproates , Cell Line, Tumor , Citric Acid , Curcumin/pharmacology , Doxorubicin/pharmacology , Drug Carriers , Drug Delivery Systems , Lactones , Particle Size , Polyesters
6.
Life Sci ; 247: 117437, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32070710

ABSTRACT

Although both the incidence and the mortality rate of breast cancer is rising, there is no potent and practical option for the treatment of these patients, particularly in advanced stages. One of the most critical challenges for treatment is the presence of complicated and extensive tumor escape mechanisms in the tumor microenvironment. Immune checkpoint molecules are of the main immunosuppressive mechanisms used by cancerous cells to block anti-cancer immune responses. Among these molecules, PD-1 (Programmed cell death) and PD-L1 (programmed cell death-ligand 1) have been considered as worthy therapeutic targets for breast cancer therapy. In this review, we intend to discuss the immunobiology and signaling of the PD-1/PD-L1 axis and highlight its importance as a worthy therapeutic target in breast cancer. We believe that the prognostic value of PD-L1 depends on the breast cancer subtype. Moreover, the combination of PD-1/PD-L1 targeting with immune-stimulating vaccines can be considered as an effective therapeutic strategy in breast cancer.


Subject(s)
Antineoplastic Agents/chemistry , B7-H1 Antigen/metabolism , Breast Neoplasms/drug therapy , Molecular Targeted Therapy/methods , Programmed Cell Death 1 Receptor/metabolism , Animals , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/chemistry , B7-H1 Antigen/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunotherapy , Programmed Cell Death 1 Receptor/chemistry , Programmed Cell Death 1 Receptor/genetics , Protein Binding/drug effects , Protein Conformation , Signal Transduction , T-Lymphocytes/drug effects , Tumor Escape/drug effects , Tumor Microenvironment/drug effects
7.
J Cell Physiol ; 235(6): 5059-5071, 2020 06.
Article in English | MEDLINE | ID: mdl-31951025

ABSTRACT

Breast cancer is a major clinical challenge that affects a wide range of the female population and heavily burdens the health system. In the past few decades, attempts have been made to understand the etiology of breast cancer, possible environmental risk factors, and the genetic predispositions, pathogenesis, and molecular aberrations involved in the process. Studies have shown that breast cancer is a heterogeneous entity; each subtype has its specific set of aberrations in different cell signaling pathways, such as Notch, Wnt/ß-catenin, transforming growth factor-ß, and mitogen-activated protein kinase pathways. One novel group of molecules that have been shown to be inducted in the regulation of multiple cell signaling pathways is the long noncoding RNAs (lncRNAs). These molecules have important implications in the regulation of multiple signaling pathways by interacting with various genes, affecting the transcription process, and finally, playing roles in posttranslational control of these genes. There is growing evidence that lncRNAs are involved in the process of breast cancer formation by effecting the aforementioned signaling pathways, and that this involvement can have significant diagnostic and prognostic values in clinical contexts. The present review aims to elicit the significance of lncRNAs in the regulation of cell signaling pathways, and the resulting changes in cell survival, proliferation, and invasion, which are the hallmarks of breast cancer.


Subject(s)
Breast Neoplasms/genetics , Cell Proliferation/genetics , RNA, Long Noncoding/genetics , Breast Neoplasms/pathology , Cell Movement/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Prognosis
8.
J Cell Physiol ; 235(6): 5008-5029, 2020 06.
Article in English | MEDLINE | ID: mdl-31724738

ABSTRACT

Breast cancer is one of the most lethal malignancies in women in the world. Various factors are involved in the development and promotion of the malignancy; most of them involve changes in the expression of certain genes, such as microRNAs (miRNAs). MiRNAs can regulate signaling pathways negatively or positively, thereby affecting tumorigenesis and various aspects of cancer progression, particularly breast cancer. Besides, accumulating data demonstrated that miRNAs are a novel tool for prognosis and diagnosis of breast cancer patients. Herein, we will review the roles of these RNA molecules in several important signaling pathways, such as transforming growth factor, Wnt, Notch, nuclear factor-κ B, phosphoinositide-3-kinase/Akt, and extracellular-signal-regulated kinase/mitogen activated protein kinase signaling pathways in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , MicroRNAs/genetics , Breast Neoplasms/pathology , Female , Humans , MAP Kinase Signaling System/genetics , NF-kappa B/genetics , Phosphatidylinositol 3-Kinase/genetics , Receptors, Notch/genetics , Signal Transduction/genetics , Wnt Proteins/genetics
9.
Bosn J Basic Med Sci ; 18(4): 328-335, 2018 Nov 07.
Article in English | MEDLINE | ID: mdl-30054999

ABSTRACT

Numerous studies indicated that microRNAs are critical in the regulation of cellular differentiation, by controlling the expression of underlying genes. The aim of this study was to investigate the effect of miR-210 upregulation on differentiation of human umbilical cord blood (HUCB)-derived mesenchymal stem cells (MSCs) into osteoblasts. MSCs were isolated from HUCB and confirmed by their adipogenic/osteogenic differentiation and flow cytometric analysis of surface markers. Pre-miR-210 was amplified from human DNA, digested and ligated with plenti-III-mir-green fluorescent protein (GFP) vector, and cloned in STBL4 bacteria. After confirmation with polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP), the plenti-III-GFP segment bearing pre-miR-210 was transfected into MSCs by electroporation. Two control vectors, pmaxGFP and Scramble, were transfected separately into MSCs. The expression of miR-210 and genes related to osteoblast differentiation, i.e., runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP) and osteocalcin gene, in the three groups of transfected MSCs was analyzed 0, 7, 14, and 21 days of transfection by quantitative reverse transcription PCR (qRT-PCR). Overexpression of miR-210 was observed in MSCs transfected with miR-210-bearing plasmid, and this was significantly different compared to Scramble group (p < 0.05). Significantly increased expression of Runx2 (at day 7 and 14), ALP and osteocalcin genes (at all time points for both genes) was observed in MSCs with miR-210-bearing plasmid compared to controls. Overall, the overexpression of miR-210 in MSCs led to MSC differentiation into osteoblasts, most probably by upregulating the Runx2, ALP, and osteocalcin genes at different stages of cell differentiation. Our study confirms the potential of miRNAs in developing novel therapeutic strategies that could target regulatory mechanisms of cellular differentiation in various disease states.


Subject(s)
Mesenchymal Stem Cells/drug effects , MicroRNAs/biosynthesis , Osteoblasts/drug effects , Cell Differentiation/drug effects , Core Binding Factor Alpha 1 Subunit/biosynthesis , Green Fluorescent Proteins , Humans , MicroRNAs/genetics , Osteocalcin/biosynthesis , Osteogenesis , Polymorphism, Restriction Fragment Length , Up-Regulation
10.
Mech Ageing Dev ; 163: 40-45, 2017 04.
Article in English | MEDLINE | ID: mdl-28043814

ABSTRACT

Autophagy is a cellular homeostatic process whereby damaged proteins and organelles are encapsulated into double membrane vesicles, called autophagosomes, for lysosomal digestion. Beclin1 plays a key role in the initial steps of autophagosome formation. In this study, we evaluated the effect of Beclin 1 overexpression in induction of autophagy and the relationship between autophagy induction and telomerase activity in HeLa cells. We found that overexpression of Beclin 1 in HeLa cells leads to autophagosome formation as shown by intracellular autophagosomal marker LC3-II staining. Expression of Beclin1 reduced telomerase activity for about 100 fold compared with the control while it did not affect TERT expression level. The results of cell cycle analysis indicated that the cell cycle and proliferation progressed normally up to 48h post-transfection. Understanding the role of autophagy induction and telomerase in the pathophysiology of aging and human cancer reveal new strategies that hold much promise for intervention and therapeutic uses.


Subject(s)
Autophagosomes/metabolism , Autophagy , Beclin-1/biosynthesis , Microtubule-Associated Proteins/metabolism , Telomerase/metabolism , Beclin-1/genetics , HeLa Cells , Humans , Microtubule-Associated Proteins/genetics , Telomerase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...