Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters











Publication year range
2.
Egypt J Immunol ; 30(4): 145-154, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37801033

ABSTRACT

Although many drugs are available for childhood systemic lupus erythematosus (SLE) treatment, the adverse effects and poor response in some cases make it crucial to find new drugs targeting various pathways in disease pathogenesis to improve overall outcomes. This study aimed to (i) investigate the effect of Panobinostat on cultured lymphocytes obtained from children with active SLE and (ii) to compare that effect with standard drugs used in SLE, such as Prednisone and hydroxychloroquine. The study included 24 SLE active patients, divided into four equal groups. Lymphocytes were isolated from blood samples of the study patients. According to the study group, cells were treated with either Panobinostat, Prednisolone, hydroxychloroquine, or not treated (control group). After cell culture, the response of lymphocytes upon drug treatment was analyzed in terms of the production of anti-dsDNA antibodies and levels of apoptosis as detected by flow cytometry using annexin V and propidium iodide (PI) staining. The Panobinostat group showed a significant decrease in the viable cell count (p < 0.001). Both Prednisone and hydroxychloroquine decreased anti-dsDNA expression more than the Panobinostat and control groups (p < 0.001 for both). PI was higher in the Prednisone group, and Annexin V was higher in the Panobinostat group compared to other groups; however, their increase did not reach statistically significant levels (p= 0.12 and 0.85, respectively). This is the first study of the Panobinostat effect on cultured lymphocytes of SLE. In conclusion, Panobinostat could be a prospective treatment for B-cell-driven autoimmune diseases such as SLE. However, its effect on autoantibodies levels and different clinical features of SLE still need a thorough evaluation.


Subject(s)
Hydroxychloroquine , Lupus Erythematosus, Systemic , Humans , Child , Hydroxychloroquine/pharmacology , Hydroxychloroquine/therapeutic use , Panobinostat/pharmacology , Panobinostat/therapeutic use , Prednisone/pharmacology , Prednisone/therapeutic use , Annexin A5/pharmacology , Annexin A5/therapeutic use , Lymphocytes
3.
Clin Transl Sci ; 16(9): 1725-1735, 2023 09.
Article in English | MEDLINE | ID: mdl-37477356

ABSTRACT

Monensin is an ionophore antibiotic that inhibits the growth of cancer cells. The aim of this study was to investigate the apoptosis-mediated anticarcinogenic effects of monensin in SH-SY5Y neuroblastoma cells. The effects of monensin on cell viability, invasion, migration, and colony formation were determined by XTT, matrigel-chamber, wound healing, and colony formation tests, respectively. The effects of monensin on apoptosis were determined by real-time polymerase chain reaction, TUNEL, Western blot, and Annexin V assay. We have shown that monensin suppresses neuroblastoma cell viability, invasion, migration, and colony formation. Moreover, we reported that monensin inhibits cell viability by triggering apoptosis of neuroblastoma cells. Monensin caused apoptosis by increasing caspase-3, 7, 8, and 9 expressions and decreasing Bax and Bcl-2 expressions in neuroblastoma cells. In Annexin V results, the rates of apoptotic cells were found to be 9.66 ± 0.01% (p < 0.001), 29.28 ± 0.88% (p < 0.01), and 62.55 ± 2.36% (p < 0.01) in the 8, 16, and 32 µM monensin groups, respectively. In TUNEL results, these values were, respectively; 35 ± 2% (p < 0.001), 34 ± 0.57% (p < 0.001), and 75 ± 2.51% (p < 0.001). Our results suggest that monensin may be a safe and effective therapeutic candidate for treating pediatric neuroblastoma.


Subject(s)
Neuroblastoma , Humans , Child , Neuroblastoma/drug therapy , Monensin/pharmacology , Monensin/therapeutic use , Annexin A5/pharmacology , Annexin A5/therapeutic use , Apoptosis , Cell Line, Tumor , Cell Proliferation
4.
Clin Exp Rheumatol ; 41(11): 2207-2215, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37083154

ABSTRACT

OBJECTIVES: Interleukin (IL)-18 plays a pro-inflammatory role in rheumatoid arthritis (RA), and its soluble inhibitor IL-18 binding protein (IL-18BP) has a potential therapeutic role. We investigated the role of IL-18BP on the joint destruction process of RA by accessing the effects of IL-18BP on fibroblast-like synoviocytes (FLSs) and chondrocytes. METHODS: Peripheral blood mononuclear cells (PBMCs) from patients with RA and healthy controls were cultured under T cell proliferative conditions with 10, 50, or 100 ng/mL of IL-18BP. After three days of culture, flow cytometry for CD4+ T cells was performed using various IL-18BP concentrations. The apoptosis and necroptosis of FLSs and chondrocytes were measured by flow cytometry using annexin V and propidium iodide (PI) and western blot under TNF-α stimulation with IL-18BP (10, 50, and 100 ng/mL). RESULTS: Differentiation of CD4+ IL-17A+ and CD4+ IL-4+ cells decreased and that of CD4+ CD25high Foxp3+ and CD4+ interferon (IFN)-γ+ cells increased on addition of IL-18BP to cultured RA patient-driven PBMCs. RA-FLS migration ability was not suppressed by IL-18BP after 12 or 24 h. IL-18BP increased annexin V+ FLS level and reduced annexin V+ chondrocyte level in a dose-dependent manner, whereas PI+ annexin V- FLS and chondrocyte levels were suppressed by 50, 100 ng/mL IL-18BP in culture. CONCLUSIONS: The administration of IL-18BP regulated the type 17 helper T cell/ regulatory T cell imbalance and attenuated the production of pro-inflammatory cytokines. IL-18BP further increased FLS apoptosis and decreased the necroptosis of FLS/chondrocytes and apoptosis of chondrocytes suggesting the joint preservative potential of IL-18BP.


Subject(s)
Arthritis, Rheumatoid , Synoviocytes , Humans , Chondrocytes/metabolism , Leukocytes, Mononuclear/metabolism , Necroptosis , Annexin A5/pharmacology , Annexin A5/metabolism , Annexin A5/therapeutic use , Arthritis, Rheumatoid/drug therapy , Cells, Cultured , Fibroblasts/metabolism , Apoptosis , Cell Proliferation
5.
Acta Biomed ; 94(2): e2023084, 2023 04 24.
Article in English | MEDLINE | ID: mdl-37092633

ABSTRACT

BACKGROUND AND AIM: VAP remains the second leading cause of death among the patients with nosocomial infections and its incidence varies significantly from 5% to 60% reaching on average 10 %. It is of crucial importance to develop novel treatment approaches and optimize the existing ones. Thus, the aim of this pilot study was to study the laboratory-microbiological effect of inhaled aminoglycosides in a complex treatment of patients with ventilator-associatedpneumonia(VAP).  Methods: To study the laboratory-microbiological effect of adjunctive inhaled aminoglycosides in the treatment of VAP, twenty enrolled patients were randomly subdivided into 2 groups (n=10). Amikacin was administered via a nebulizer starting from the first day of VAP manifestation. Inhalations were performed BID for 7 days via a nebulizer integrated into the breathing circuit. We assessed: cell membrane alterations in leukocytes, Annexin V/7-AAD staining for leukocytes, ROS detection assay for leukocytes. RESULTS: Adjunctive administration of inhaled amikacin reduced the fluorescence intensity ratio more efficiently compared with the intravenous antimicrobial treatment with no aerosolized amikacin following both 48 h and 96 h of treatment. The amount of dead necrotic annexin V-negative, 7-AAD-positive leukocytes was significantly lower under the use of inhaled amikacin than at the beginning of treatment. Conclusions In this pilot study, we found that administration of aerosolized amikacin combined with the systemic antimicrobial therapy improves the clinical outcome of patients with VAP, effective early microbial decrease in the sputum, reduces reactive oxygen species generation in leukocytes and the degree of leukocyte apoptosis and necrosis, decreases VAP-mediated cell membrane alterations of circulating leukocytes.


Subject(s)
Amikacin , Pneumonia, Ventilator-Associated , Humans , Infant , Amikacin/therapeutic use , Amikacin/adverse effects , Pneumonia, Ventilator-Associated/etiology , Pneumonia, Ventilator-Associated/microbiology , Pilot Projects , Annexin A5/therapeutic use , Administration, Inhalation , Anti-Bacterial Agents/adverse effects , Aminoglycosides
6.
Int J Mol Sci ; 24(4)2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36835282

ABSTRACT

Malignant melanoma, an increasingly common form of skin cancer, is a major threat to public health, especially when the disease progresses past skin lesions to the stage of advanced metastasis. Targeted drug development is an effective strategy for the treatment of malignant melanoma. In this work, a new antimelanoma tumor peptide, the lebestatin-annexin V (designated LbtA5) fusion protein, was developed and synthesized by recombinant DNA techniques. As a control, annexin V (designated ANV) was also synthesized by the same method. The fusion protein combines annexin V, which specifically recognizes and binds phosphatidylserine, with the disintegrin lebestatin (lbt), a polypeptide that specifically recognizes and binds integrin α1ß1. LbtA5 was successfully prepared with good stability and high purity while retaining the dual biological activity of ANV and lbt. MTT assays demonstrated that both ANV and LbtA5 could reduce the viability of melanoma B16F10 cells, but the activity of the fusion protein LbtA5 was superior to that of ANV. The tumor volume growth was slowed in a mouse xenograft model treated with ANV and LbtA5, and the inhibitory effect of high concentrations of LbtA5 was significantly better than that of the same dose of ANV and was comparable to that of DTIC, a drug used clinically for melanoma treatment. The hematoxylin and eosin (H&E) staining test showed that ANV and LbtA5 had antitumor effects, but LbtA5 showed a stronger ability to induce melanoma necrosis in mice. Immunohistochemical experiments further showed that ANV and LbtA5 may inhibit tumor growth by inhibiting angiogenesis in tumor tissue. Fluorescence labeling experiments showed that the fusion of ANV with lbt enhanced the targeting of LbtA5 to mouse melanoma tumor tissue, and the amount of target protein in tumor tissue was significantly increased. In conclusion, effective coupling of the integrin α1ß1-specific recognition molecule lbt confers stronger biological antimelanoma effects of ANV, which may be achieved by the dual effects of effective inhibition of B16F10 melanoma cell viability and inhibition of tumor tissue angiogenesis. The present study describes a new potential strategy for the application of the promising recombinant fusion protein LbtA5 in the treatment of various cancers, including malignant melanoma.


Subject(s)
Annexin A5 , Integrin alpha1beta1 , Melanoma , Recombinant Fusion Proteins , Skin Neoplasms , Animals , Humans , Mice , Annexin A5/therapeutic use , Integrin alpha1beta1/metabolism , Melanoma/therapy , Recombinant Fusion Proteins/therapeutic use , Skin Neoplasms/therapy , Melanoma, Experimental , Melanoma, Cutaneous Malignant
7.
Folia Biol (Praha) ; 68(1): 16-24, 2022.
Article in English | MEDLINE | ID: mdl-36201854

ABSTRACT

Non-small cell lung cancer (NSCLC) results in high mortality and has gained increasing attention. C-Phycocyanin (C-PC) has been identified as a potential therapeutic inhibitor for NSCLC, but its underlying mechanism remains obscure. The gene expression of the long noncoding RNA neighbour of BRCAI RNA 2 (NBR2) in NSCLC cells was evaluated by quantitative reverse transcription-PCR. The cell capacity for proliferation and migration was examined by EdU and wound-healing assays. Furthermore, the viability and apoptosis of cells was measured with CCK-8 and annexin V/PI, respectively. Next, the protein level of activation of adenosine monophosphate- activated protein kinase and the rapamycin kinase (mTOR) signalling pathway-associated molecules was evaluated by western blotting. H292 cells were pre-treated with C-PC or transfected with plasmids encoding NBR2 or the shNBR2 plasmid, to over-express or knock down NBR2 expression, respectively. NBR2 expression was robustly down-regulated in NSCLC cell lines compared with a normal cell line (BEAS-2B). NBR2 over-expression inhibited migration and promoted apoptosis of H292 cells. Treatment of H292 cells with C-PC enhanced NBR2 levels in a dose- and time-dependent manner. Downregulation of NBR2 in H292 cells inhibited the activity of C-PC on cell proliferation, viability and clone formation. Further mechanistic investigation showed that the down-regulation of NBR2 abolished the modulatory effects of C-PC on the AMPK/mTOR signalling pathway. In conclusion, C-PC inhibits H292 cell growth by enhancing the NBR2/AMPK signalling pathway.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , RNA, Long Noncoding , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/pharmacology , Adenosine Monophosphate/pharmacology , Adenosine Monophosphate/therapeutic use , Annexin A5/pharmacology , Annexin A5/therapeutic use , Apoptosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Phycocyanin/metabolism , Phycocyanin/pharmacology , Phycocyanin/therapeutic use , RNA, Long Noncoding/genetics , Sincalide/metabolism , Sincalide/pharmacology , Sincalide/therapeutic use , Sirolimus/pharmacology , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/metabolism
8.
J Ethnopharmacol ; 299: 115658, 2022 Dec 05.
Article in English | MEDLINE | ID: mdl-36075273

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Artemisia argyi H. Lév. & Vaniot (Asteraceae), also called "Chinese mugwort", is frequently used as a herbal medicine in China, Japan, Korea, and eastern parts of Russia. It is known as "ai ye" in China and "Gaiyou" in Japan. In ancient China, the buds and leaves of A. argyi were commonly consumed before and after Tomb-sweeping Day. It is used to treat malaria, hepatitis, cancer, inflammatory diseases, asthma, irregular menstrual cycle, sinusitis, and pathologic conditions of the kidney and liver. Although A. argyi extract (AAE) has shown anti-tumor activity against various cancers, the therapeutic effect and molecular mechanism of AAE remains to be further studied in lung cancer. AIM OF THE STUDY: This study aimed to demonstrate the anti-tumor effect of AAE and its associated biological mechanisms in CL1-0 parent and gemcitabine-resistant (CL1-0-GR) lung cancer cells. EXPERIMENTAL PROCEDURE: Human lung cancer cells CL1-0 and CL1-0-GR cells were treated with AAE. Cell viability was assessed using the MTT, colony, and spheroid formation assays. Migration, invasion, and immunofluorescence staining were used to determine the extent of epithelial- mesenchymal transition (EMT). JC-1 and MitoSOX fluorescent assays were performed to investigate the effect of AAE on mitochondria. Apoptosis was detected using the TUNEL assay and flow cytometry with Annexin V staining. RESULT: We found that A. argyi significantly decreased cell viability and induced apoptosis, accompanied by mitochondrial membrane depolarization and increased ROS levels in both parent cells (CL1-0) and gemcitabine-resistant lung cancer cells (CL1-0-GR). AAE-induced apoptosis is regulated via the PI3K/AKT and MAPK signaling pathways. It also prevents CL1-0 and CL1-0-GR cancer cell invasion, migration, EMT, colony formation, and spheroid formation. In addition, AAE acts cooperative with commercial chemotherapy drugs to enhance tumor spheroid shrinkage. CONCLUSION: Our study provides the first evidence that A. argyi treatment suppresses both parent and gemcitabine-resistant lung cancer cells by inducing ROS, mitochondrial membrane depolarization, and apoptosis, and reducing EMT. Our finding provides insights into the anti-cancer activity of A. argyi and suggests that A. argyi may serve as a chemotherapy adjuvant that potentiates the efficacy of chemotherapeutic agents.


Subject(s)
Apoptosis , Artemisia , Lung Neoplasms , Annexin A5/metabolism , Annexin A5/pharmacology , Annexin A5/therapeutic use , Apoptosis/drug effects , Artemisia/chemistry , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Humans , Lung Neoplasms/drug therapy , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Gemcitabine
9.
Neoplasma ; 69(5): 1119-1128, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36004650

ABSTRACT

Prostate cancer is one of the most frequently diagnosed cancers in men. The medical treatment of metastatic prostate cancer relies heavily on androgen deprivation. The present study aimed to explore the inhibitory effect of anlotinib on androgen receptor (AR)-negative prostate cancer cell lines in vitro and investigate its mechanism of action. Two prostate cancer cell lines, DU145 and PC-3, were treated with different concentrations (0-80 µM) of anlotinib. Cell proliferation was accessed by CCK-8 assay and EdU staining. Cell nuclear morphology was observed after DAPI staining, cell apoptosis level was evaluated by Annexin-V-FITC/PI staining, and western blot was used to detect the proliferation- and apoptosis-related proteins. The potential interaction between anlotinib and AKT was revealed by molecular docking. After treatment with anlotinib, the cell proliferation rate was significantly inhibited in a dose-dependent manner. The DAPI staining showed that anlotinib could induce apoptosis. Further, Annexin V/PI double staining confirmed the occurrence of apoptosis, accompanied by the increase of cleaved caspase-3 and activated PARP. Moreover, anlotinib significantly decreased the phosphorylation of protein kinase B (AKT) and its downstream pathway proteins in prostate cells (p<0.05). Experiments further confirmed that the activation of the AKT pathway reversed the inhibitory effect of anlotinib on DU145 and PC-3 cell proliferation. In addition, molecular docking analysis revealed potential interactions between anlotinib and AKT1 at multiple sites. Overall, the present study suggested that anlotinib could inhibit the proliferation and induce apoptosis in the AR-negative prostate cancer cell lines, possibly via the inactivation of the AKT pathway.


Subject(s)
Prostatic Neoplasms , Proto-Oncogene Proteins c-akt , Androgen Antagonists/therapeutic use , Androgens/pharmacology , Annexin A5/metabolism , Annexin A5/pharmacology , Annexin A5/therapeutic use , Apoptosis , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation , Fluorescein-5-isothiocyanate/pharmacology , Fluorescein-5-isothiocyanate/therapeutic use , Humans , Indoles , Male , Molecular Docking Simulation , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Prostate/pathology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Quinolines , Receptors, Androgen , Signal Transduction
10.
Pharmacol Rep ; 74(5): 998-1010, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35908023

ABSTRACT

BACKGROUND: The lack of drug targets is an obstacle to the treatment of patients with triple-negative breast cancer (TNBC). At present, non-specific cytotoxic drugs are first-line agents, but the development of resistance is a major problem with these agents. The epidermal growth factor receptor (EGFR) is a potential target in some TNBCs, because its tyrosine kinase activity drives tumorigenesis. Thus, small molecule inhibitors of the EGFR in combination with cytotoxic agents could be important for the treatment of TNBCs. METHODS: The present study evaluated the efficacies of clinically approved EGFR inhibitors in combination with the cytotoxic agent ixabepilone in parental and docetaxel-resistant MDA-MB-231 cells (231C and TXT cells, respectively). Cell viability was assessed using MTT reduction assays, cell death pathways were evaluated using annexin V/7-aminoactinomycin D staining and flow cytometry and Western immunoblotting was used to assess the expression of pro- and anti-apoptotic proteins in cells. RESULTS: Ixabepilone and the EGFR inhibitors gefitinib and vandetanib inhibited 231C and TXT cell proliferation, but the alternate EGFR inhibitors erlotinib and lapatinib were poorly active. Using combination analysis, ixabepilone/vandetanib was synergistic in both cell types, whereas the ixabepilone/gefitinib combination exhibited antagonism. By flow cytometry, ixabepilone/vandetanib enhanced 231C and TXT cell death over that produced by the single agents and also enhanced caspase-3 cleavage and the pro/anti-apoptotic Bcl-2 protein ratios over ixabepilone alone. CONCLUSIONS: These findings suggest that the ixabepilone/vandetanib combination may have promise for the treatment of patients with drug-resistant TNBC.


Subject(s)
Antineoplastic Agents , Triple Negative Breast Neoplasms , Humans , Docetaxel/pharmacology , Gefitinib/pharmacology , Gefitinib/therapeutic use , Lapatinib/pharmacology , Caspase 3/metabolism , Erlotinib Hydrochloride/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Annexin A5/pharmacology , Annexin A5/therapeutic use , ErbB Receptors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Protein Kinase Inhibitors/pharmacology , Cell Proliferation , Proto-Oncogene Proteins c-bcl-2/metabolism , Cytotoxins/pharmacology , Cell Line, Tumor , Apoptosis
11.
Adv Healthc Mater ; 11(16): e2200416, 2022 08.
Article in English | MEDLINE | ID: mdl-35708176

ABSTRACT

Thromboembolic stroke is typically characterized by the activation of platelets, resulting in thrombus in the cerebral vascular system, leading to high morbidity and mortality globally. Intravenous thrombolysis by tissue plasminogen activator (tPA) administration within 4.5 h from the onset of symptoms is providing a standard therapeutic strategy for ischemic stroke, but this reagent simultaneously shows potential serious adverse effects, e.g., hemorrhagic transformation. Herein, a novel delivery platform based on Annexin V and platelet membrane is developed for tPA (APLT-PA) to enhance targeting efficiency, therapeutic effects, and reduce the risk of intracerebral hemorrhage in acute ischemic stroke. After preparation by extrusion of platelet membrane and subsequent insertion of Annexin V to liposomes, APLT-PA exhibits a high targeting efficiency to activated platelet in vitro and thrombosis site in vivo, due to the binding to phosphatidylserine (PS) and activated platelet membrane proteins. One dose of APLT-PA leads to obvious thrombolysis and significant improvement of neurological function within 7 days in mice with photochemically induced acute ischemic stroke. This study provides a novel, safe platelet-biomimetic nanomedicine for precise thrombolytic treatment of acute ischemic stroke, and offers new theories for the design and exploitation of cell-mimetic nanomedicine for diverse biomedical applications.


Subject(s)
Ischemic Stroke , Stroke , Animals , Annexin A5/therapeutic use , Biomimetics , Blood Platelets , Fibrinolytic Agents , Mice , Nanomedicine , Stroke/drug therapy , Thrombolytic Therapy/adverse effects , Tissue Plasminogen Activator/adverse effects
12.
Ultrastruct Pathol ; 46(4): 348-358, 2022 Jul 04.
Article in English | MEDLINE | ID: mdl-35727696

ABSTRACT

Breast cancer is the most common cancer in women in the world. Many anticancer drugs are currently used clinically have been isolated from plant species or are based on such substances. Linalool is aromatic compounds from the monoterpene group. It is the main constituents of essential oils and show antiproliferative, antioxidant, and antiseptic properties. The aim of this study was to investigate the antiproliferativeand apoptotic, effects of linalool in MCF-7 and MDA-MB-231 human breast cancer cells. MCF-7 and MDA-MB-231 human breast cancer cells were treated with different concentrations of linalool (100, 200, 400, 600, 800, 1000 µM) at 24 h and 48 h. MTT assay for cell proliferation and Annexin V assay for apoptosis was done. The morphology of breast cancer cells was investigated by light microscope and scanning electron microscope (SEM). The study show that linalool significantly induced apoptosis in all groups as dose and time-dependent (p < .05). Linalool has apoptotic and antiproliferative properties in a concentration and time-dependent manner in breast cancer cells. The cytotoxic effects of linalool on MCF-7 and MDA-MB-231 human breast cancer cells was found to be associated with apoptotic cell death. Linalool was more effective on MCF-7 human breast cancer cells in smaller amounts.


Subject(s)
Anti-Infective Agents, Local , Antineoplastic Agents , Breast Neoplasms , Oils, Volatile , Acyclic Monoterpenes , Annexin A5/pharmacology , Annexin A5/therapeutic use , Anti-Infective Agents, Local/pharmacology , Anti-Infective Agents, Local/therapeutic use , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Apoptosis , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Female , Humans , MCF-7 Cells , Monoterpenes/pharmacology , Monoterpenes/therapeutic use , Oils, Volatile/pharmacology
13.
Mol Pharm ; 19(7): 2429-2440, 2022 07 04.
Article in English | MEDLINE | ID: mdl-35639628

ABSTRACT

In breast cancer therapy, Gemcitabine (Gem) is an antineoplastic antimetabolite with greater anticancer efficacy and tolerability. However, effectiveness of Gem is limited by its off-target effects. The synergistic potential of MUC1 (mucin 1) inhibitors and Gem-loaded polymeric nanoparticles (NPs) was discussed in this work in order to reduce dose-related toxicities and enhance the therapeutic efficacy. The double emulsion solvent evaporation method was used to prepare poly(ethylene glycol) methyl ether-block-poly-caprolactone (PEG-PCL)-loaded Gem and MUC 1 inhibitor NPs. The average size of Gem and MUC 1 inhibitor-loaded NPs was 128 nm, with a spherical shape. Twin-loaded NPs containing Gem and the MUC1 inhibitor decreased IC50 and behaved synergistically. Furthermore, in vitro mechanistic studies, that is, loss of MMP, clonogenic assay, Annexin V FITC assay, and Western blotting to confirm apoptosis with simultaneous induction of autophagy using acridine orange (AO) staining were performed in this study. Furthermore, the investigated NPs upon combination exhibited greater loss of MMP and decreased clonogenic potential with simultaneous induction of autophagy in MCF-7 cells. Annexin V FITC clearly showed the percentage of apoptosis while Western blotting protein expression analysis revealed an increase in caspase-3 activity with simultaneous decrease in Bcl-2 protein expression, a hallmark of apoptosis. The effectiveness of the Ehrlich ascites solid (EAT) mice treated with Gem-MUC1 inhibitor NPs was higher than that of the animals treated alone. Overall, the combined administration of Gem and MUC1 inhibitor-loaded NPs was found to be more efficacious than Gem and MUC1 inhibitor delivered separately.


Subject(s)
Breast Neoplasms , Nanoparticles , Animals , Annexin A5/therapeutic use , Breast Neoplasms/drug therapy , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Female , Fluorescein-5-isothiocyanate , Humans , Mice , Mucin-1 , Polyesters , Polyethylene Glycols , Gemcitabine
14.
Biol Trace Elem Res ; 200(11): 4807-4816, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35508889

ABSTRACT

Obesity is a chronic disease associated with increased morbidity and mortality. The rapidly increasing prevalence of obesity makes it a global health problem, while treatment options remain limited. Given the potential of boron in the treatment of obesity, the aim of this study is to investigate the anti-adipogenic activity of the newly synthesised boron glycine monoester compound (BGM) using 3T3-L1 adipocytes by analysing lipid accumulation, CTRP3 and PPARy gene expression, oxidative stress and apoptotic effects. 3T3-L1 fibroblast cells (ATCC® CL-173) were transformed into adipocyte cells in vitro. Fat accumulation in the 3T3-L1 adipocyte cells was detected by Oil Red O staining. Gene expression levels were determined with qPCR. Biochemical analyzes were performed using spectrophotometric method (CAT, ALP and ACP) and ELISA kit (TAS, TOS, NADP-IDH). Apoptosis studies were performed on the muse cell nalyser using the Muse Annexin V & Dead Cell Assay Kit. When BGM-treated cells were compared to control adipocyte cells, lipid accumulation decreased in a dose-dependent manner. BGM-treated adipocyte cells had higher CTRP3 expression levels and lower PPAR-γ gene expression levels compared to control adipocyte cells (p < 0.001). While BGM application increased the TAS level, it showed an antioxidant effect by regulating the activity of oxidative metabolism enzymes (p < 0.001). BGM application increased total apoptosis by 1.5-fold. These results show that BGM is a potential therapeutic agent for obesity by regulating the expression of genes related to adipogenesis and lipogenesis in adipocyte cells and by affecting the activity of enzymes of oxidative metabolism and apoptosis.


Subject(s)
Boron , PPAR gamma , 3T3-L1 Cells , Adipocytes , Adipogenesis , Alprostadil/metabolism , Alprostadil/pharmacology , Alprostadil/therapeutic use , Animals , Annexin A5/metabolism , Annexin A5/pharmacology , Annexin A5/therapeutic use , Antioxidants/metabolism , Apoptosis , Boron/pharmacology , Cell Differentiation , Glycine/pharmacology , Lipogenesis , Mice , NADP/metabolism , NADP/pharmacology , NADP/therapeutic use , Obesity/metabolism , Oxidative Stress , PPAR gamma/genetics , PPAR gamma/metabolism
15.
Blood ; 138(25): 2714-2726, 2021 12 23.
Article in English | MEDLINE | ID: mdl-34610086

ABSTRACT

Severe traumatic brain injury (TBI) often causes an acute systemic hypercoagulable state that rapidly develops into consumptive coagulopathy. We have recently demonstrated that TBI-induced coagulopathy (TBI-IC) is initiated and disseminated by brain-derived extracellular vesicles (BDEVs) and propagated by extracellular vesicles (EVs) from endothelial cells and platelets. Here, we present results from a study designed to test the hypothesis that anticoagulation targeting anionic phospholipid-expressing EVs prevents TBI-IC and improves the outcomes of mice subjected to severe TBI. We evaluated the effects of a fusion protein (ANV-6L15) for improving the outcomes of TBI in mouse models combined with in vitro experiments. ANV-6L15 combines the phosphatidylserine (PS)-binding annexin V (ANV) with a peptide anticoagulant modified to preferentially target extrinsic coagulation. We found that ANV-6L15 reduced intracranial hematoma by 70.2%, improved neurological function, and reduced death by 56.8% in mice subjected to fluid percussion injury at 1.9 atm. It protected the TBI mice by preventing vascular leakage, tissue edema, and the TBI-induced hypercoagulable state. We further showed that the extrinsic tenase complex was formed on the surfaces of circulating EVs, with the highest level found on BDEVs. The phospholipidomic analysis detected the highest levels of PS on BDEVs, as compared with EVs from endothelial cells and platelets (79.1, 15.2, and 3.5 nM/mg of protein, respectively). These findings demonstrate that TBI-IC results from a trauma-induced hypercoagulable state and may be treated by anticoagulation targeting on the anionic phospholipid-expressing membrane of EVs from the brain and other cells.


Subject(s)
Annexin A5/therapeutic use , Anticoagulants/therapeutic use , Brain Injuries, Traumatic/drug therapy , Extracellular Vesicles/drug effects , Phospholipids/metabolism , Recombinant Fusion Proteins/therapeutic use , Thrombophilia/drug therapy , Animals , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Extracellular Vesicles/metabolism , Extracellular Vesicles/pathology , Male , Mice, Inbred C57BL , Thrombophilia/etiology , Thrombophilia/metabolism , Thrombophilia/pathology
16.
Mediators Inflamm ; 2021: 1946384, 2021.
Article in English | MEDLINE | ID: mdl-33927569

ABSTRACT

BACKGROUND: The recombinant protein diannexin can inhibit platelet-mediated events, which contribute to acute respiratory distress syndrome (ARDS). Here, we investigated the effect of diannexin and its effect on heme oxygenase-1 (HO-1) in ARDS. METHODS: A total of 32 rats were randomized into sham, ARDS, diannexin (D), and diannexin+HO-1 inhibitor (DH) groups. Alveolar-capillary permeability was evaluated by testing the partial pressure of oxygen to fraction of inspired oxygen (PaO2/FiO2) ratio, lung wet/dry weight ratio, and protein levels in the lung. Inflammation was assessed by measuring cytokine levels in the bronchial alveolar lavage fluid (BALF) and serum and nuclear factor-κB (NF-κB) in the lung tissue. Inducible nitric oxide synthase (iNOS), malondialdehyde (MDA), and myeloperoxidase (MPO) were measured to evaluate the oxidative stress response. Lung tissue pathology and apoptosis were also evaluated. We measured HO-1 expression in the lung tissue to investigate the effect of diannexin on HO-1 in ARDS. RESULTS: Compared with the ARDS group, diannexin improved PaO2/FiO2, lung wet/dry weight ratio, and protein levels in the BALF and decreased levels of cytokines and NF-κB in the lung and serum. Diannexin inhibited the oxidative stress response and significantly ameliorated pathological lung injury and apoptosis. The partial reversal of diannexin effects by a HO-1 inhibitor suggests that diannexin may promote HO-1 expression to ameliorate ARDS. CONCLUSIONS: We showed that diannexin can improve alveolar-capillary permeability, inhibit the oxidative stress response and inflammation, and protect against ARDS-induced lung injury and apoptosis.


Subject(s)
Annexin A5/therapeutic use , Heme Oxygenase-1/physiology , Respiratory Distress Syndrome/drug therapy , Animals , Annexin A5/pharmacology , Apoptosis/drug effects , Blood Coagulation/drug effects , Capillary Permeability/drug effects , Heme Oxygenase-1/genetics , Inflammation/prevention & control , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Respiratory Distress Syndrome/metabolism
17.
Nat Commun ; 11(1): 1137, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32111835

ABSTRACT

The interaction between immune cells and phosphatidylserine (PS) molecules exposed on the surface of apoptotic-tumor bodies, such as those induced by chemotherapies, contributes to the formation of an immunosuppressive tumor microenvironment (TME). Annexin A5 (AnxA5) binds with high affinity to PS externalized by apoptotic cells, thereby hindering their interaction with immune cells. Here, we show that AnxA5 administration rescue the immunosuppressive state of the TME induced by chemotherapy. Due to the preferential homing of AnxA5 to the TME enriched with PS+ tumor cells, we demonstrate in vivo that fusing tumor-antigen peptide to AnxA5 significantly enhances its immunogenicity and antitumor efficacy when administered after chemotherapy. Also, the therapeutic antitumor effect of an AnxA5-peptide fusion can be further enhanced by administration of other immune checkpoint inhibitors. Our findings support the administration of AnxA5 following chemotherapy as a promising immune checkpoint inhibitor for cancer treatment.


Subject(s)
Annexin A5/therapeutic use , Cancer Vaccines/therapeutic use , Immunologic Factors/therapeutic use , Neoplasms/therapy , Animals , Annexin A5/genetics , Annexin A5/metabolism , Antibodies, Blocking/therapeutic use , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/therapeutic use , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cancer Vaccines/metabolism , Cell Line, Tumor , Cisplatin/adverse effects , Cisplatin/therapeutic use , Disease Models, Animal , Female , Humans , Immunologic Factors/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/immunology , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/immunology , Papillomavirus E7 Proteins/therapeutic use , Phosphatidylserines/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
18.
Med Hypotheses ; 135: 109462, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31731057

ABSTRACT

The phosphatidylserine (PS) molecule is present in cell membranes where it is actively kept on their inner leaflets but when cells are damaged it moves to the surface and become a signal for their removal, the platform upon which the coagulation cascade takes place and a ligand that activates a feedback cycle of inflammatory cytokine secretion and initiates the wakeup call for the innate immune response. These are physiologic responses to PS but the Ebola virus displays PS molecules on its membrane's surface and the huge numbers of viruses cause a pathologic inflammatory cytokine storm and a hemorrhagic consumptive coagulopathy. Annexin V is an innate molecule that can cloak membrane displayed PS and prevents its Th1 cell's inflammatory cytokine generation and cascade thrombin generation. The hypothesis presented is that its administration will cloak PS and prevent Ebola's consumptive coagulopathy and its cytokine storm.


Subject(s)
Annexin A5/therapeutic use , Cytokines/metabolism , Disseminated Intravascular Coagulation/drug therapy , Disseminated Intravascular Coagulation/metabolism , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/metabolism , Phosphatidylserines/metabolism , Annexin A5/metabolism , Blood Coagulation , Cell Membrane/metabolism , Cytokine Release Syndrome/virology , Humans , Immunity, Innate , Inflammation , Th1 Cells/metabolism , Thrombin/metabolism
19.
Thromb Haemost ; 118(1): 90-102, 2018 01.
Article in English | MEDLINE | ID: mdl-29304529

ABSTRACT

Without conjunctive administration of an anticoagulant, endothelial injury-induced thrombosis is resistant to thrombolysis and prone to re-thrombosis. We hypothesized that co-delivery of recombinant tissue plasminogen activator (rtPA) with annexin V-containing anticoagulants that specifically target the injured endothelium may passivate the thrombogenic elements of the vascular injury site and enhance rtPA-induced thrombolysis. In this study, the effects of conjunctive administration of Kinexins (Kunitz inhibitor-annexin V fusion proteins) with rtPA on thrombolysis were determined in vitro and in vivo. Thromboelastometry showed that both TAP-A (tick anticoagulant peptide-annexin V fusion protein; an inhibitor of factor Xa [FXa] and prothrombinase) and A-6L15 (annexin V-6L15 fusion protein; an inhibitor of tissue factor/FVIIa) exerted concentration-dependent (10-100 nM) effects on clot formation, with TAP-A being several folds more potent than A-6L15 in whole blood. Combination of TAP-A or A-6L15 with rtPA (1 µg/mL) led to decrease in lysis index, suggesting conjunctive enhancement of thrombolysis by combined use of rtPA with TAP-A or A-6L15. In a rat cremaster muscle preparation subjected to photochemical injury, conjunctive administration of rtPA and TAP-A significantly restored tissue perfusion to 56%, which is approximately two fold of that by rtPA or TAP-A alone. Near-infrared fluorescence images demonstrated local retention of a fluorescent A-6L15-S288 at the injury site, suggesting a targeting effect of the fusion protein. Pharmacokinetic analysis showed that 123I-labelled TAP-A and A-6L15 had initial distribution half-lives (T1/2α) of approximately 6 minutes and elimination half-lives (T1/2ß) of approximately 2.3 hours. In conclusion, Kinexins were potentially useful adjunctive agents with rtPA thrombolytic therapy especially for thrombosis induced by endothelial injury.


Subject(s)
Abdominal Muscles/physiopathology , Annexin A5/chemistry , Endothelium/injuries , Thrombelastography , Thrombolytic Therapy , Trypsin Inhibitor, Kunitz Soybean/chemistry , Animals , Annexin A5/therapeutic use , Anticoagulants/chemistry , Blood Coagulation/drug effects , Endothelium/pathology , Factor VIIa/chemistry , Factor Xa/chemistry , Humans , Iodine Radioisotopes/chemistry , Male , Peptides/chemistry , Perfusion , Protease Inhibitors/metabolism , Rats , Rats, Sprague-Dawley , Spectrometry, Fluorescence , Spectroscopy, Near-Infrared , Thrombosis/physiopathology , Trypsin Inhibitor, Kunitz Soybean/therapeutic use
20.
Exp Biol Med (Maywood) ; 241(16): 1751-6, 2016 10.
Article in English | MEDLINE | ID: mdl-27190270

ABSTRACT

Current flow cytometry (FCM) requires fluorescent dyes labeling cells which make the procedure costly and time consuming. This manuscript reports a feasibility study of detecting the cell apoptosis with a label-free method in glioblastoma cells. A human glioma cell line M059K was exposed to 8 Gy dose of radiation, which enables the cells to undergo radiation-induced apoptosis. The rates of apoptosis were studied at different time points post-irradiation with two different methods: FCM in combination with Annexin V-FITC/PI staining and a newly developed technique named polarization diffraction imaging flow cytometry. Totally 1000 diffraction images were acquired for each sample and the gray level co-occurrence matrix (GLCM) algorithm was used in morphological characterization of the apoptotic cells. Among the feature parameters extracted from each image pair, we found that the two GLCM parameters of angular second moment (ASM) and sum entropy (SumEnt) exhibit high sensitivities and consistencies as the apoptotic rates (Pa) measured with FCM method. In addition, no significant difference exists between Pa and ASM_S, Pa and SumEnt_S, respectively (P > 0.05). These results demonstrated that the new label-free method can detect cell apoptosis effectively. Cells can be directly used in the subsequent biochemical experiments as the structure and function of cells and biomolecules are well-preserved with this new method.


Subject(s)
Apoptosis/radiation effects , Glioblastoma/radiotherapy , Annexin A5/therapeutic use , Cell Line, Tumor , Coloring Agents/therapeutic use , Feasibility Studies , Flow Cytometry/methods , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/therapeutic use , Humans , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL