Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Sci Rep ; 12(1): 8858, 2022 05 25.
Article in English | MEDLINE | ID: mdl-35614109

ABSTRACT

Apigenin is a dietary polyphenol found abundantly in fruit and vegetables, which sensitizes leukaemia cells to topoisomerase inhibitor agents (e.g., etoposide), and alkylating agents (e.g., cyclophosphamide), reducing ATP levels and inducing apoptosis; whilst being protective to control haematopoietic stem cells. This study analysed the expression profiles of intrinsic and extrinsic apoptosis-related genes and proteins to help elucidate the mechanisms of action of apigenin when used in combination with etoposide or cyclophosphamide in lymphoid and myeloid leukaemia cell lines (Jurkat and THP-1). Expression of apoptosis-related genes were measured using a TaqMan® Human Apoptosis Array and the StepOne Plus RT-qPCR System, whilst apoptosis-related proteins were determined using a protein profiler™-human apoptosis array and the LI-COR OdysseyR Infrared Imaging System. Apigenin when combined with etoposide or cyclophosphamide-induced apoptosis via the mitochondrial pathway, increasing the expression of pro-apoptotic cytochrome c, SMAC/DIABLO, and HTRA2/OMI, which promoted caspase-9 and -3 activation. Targeting anti-apoptotic and/or pro-apoptotic members of the apoptotic pathways is a promising strategy to induce cancer cell death and improve sensitivity to chemotherapy agents. Here the apoptotic pathways induced by apigenin in combination with etoposide or cyclophosphamide were identified within human leukaemia cell lines, such applications could provide combination therapies for the treatment of leukaemia.


Subject(s)
Apigenin , Apoptosis Regulatory Proteins , Apoptosis , Leukemia , Apigenin/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/genetics , Cell Line/drug effects , Cell Line/metabolism , Cyclophosphamide/pharmacology , Drug Therapy, Combination , Etoposide/pharmacology , Humans , Leukemia/drug therapy , Leukemia/genetics , Mitochondrial Proteins/metabolism
2.
Int J Mol Sci ; 22(6)2021 Mar 17.
Article in English | MEDLINE | ID: mdl-33802972

ABSTRACT

Leukaemia is a malignant disease of the blood. Current treatments for leukaemia are associated with serious side-effects. Plant-derived polyphenols have been identified as potent anti-cancer agents and have been shown to work synergistically with standard chemotherapy agents in leukaemia cell lines. Polyphenols have multiple mechanisms of action and have been reported to decrease cell proliferation, arrest cell cycle and induce apoptosis via the activation of caspase (3, 8 and 9); the loss of mitochondrial membrane potential and the release of cytochrome c. Polyphenols have been shown to suppress activation of transcription factors, including NF-kB and STAT3. Furthermore, polyphenols have pro-oxidant properties, with increasing evidence that polyphenols inhibit the antioxidant activity of glutathione, causing oxidative DNA damage. Polyphenols also induce autophagy-driven cancer cell death and regulate multidrug resistance proteins, and thus may be able to reverse resistance to chemotherapy agents. This review examines the molecular mechanism of action of polyphenols and discusses their potential therapeutic targets. Here, we discuss the pharmacological properties of polyphenols, including their anti-inflammatory, antioxidant, anti-proliferative, and anti-tumour activities, and suggest that polyphenols are potent natural agents that can be useful therapeutically; and discuss why data on bioavailability, toxicity and metabolism are essential to evaluate their clinical use.


Subject(s)
Leukemia/drug therapy , Polyphenols/therapeutic use , Animals , Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Humans , Leukemia/pathology , Models, Biological , Polyphenols/chemistry , Polyphenols/pharmacology , Reactive Oxygen Species/metabolism
3.
Front Immunol ; 11: 568931, 2020.
Article in English | MEDLINE | ID: mdl-33193345

ABSTRACT

Programmed death-ligand 1 (PD-L1) is an immune checkpoint inhibitor that binds to its receptor PD-1 expressed by T cells and other immune cells to regulate immune responses; ultimately preventing exacerbated activation and autoimmunity. Many tumors exploit this mechanism by overexpressing PD-L1 which often correlates with poor prognosis. Some tumors have also recently been shown to express PD-1. On tumors, PD-L1 binding to PD-1 on immune cells promotes immune evasion and tumor progression, primarily by inhibition of cytotoxic T lymphocyte effector function. PD-1/PD-L1-targeted therapy has revolutionized the cancer therapy landscape and has become the first-line treatment for some cancers, due to their ability to promote durable anti-tumor immune responses in select patients with advanced cancers. Despite this clinical success, some patients have shown to be unresponsive, hyperprogressive or develop resistance to PD-1/PD-L1-targeted therapy. The exact mechanisms for this are still unclear. This review will discuss the current status of PD-1/PD-L1-targeted therapy, oncogenic expression of PD-L1, the new and emerging tumor-intrinisic roles of PD-L1 and its receptor PD-1 and how they may contribute to tumor progression and immunotherapy responses as shown in different oncology models.


Subject(s)
B7-H1 Antigen/metabolism , Immune Checkpoint Proteins , Programmed Cell Death 1 Receptor/metabolism , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/genetics , Biomarkers, Tumor , Energy Metabolism , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunomodulation/drug effects , Immunomodulation/genetics , Molecular Targeted Therapy , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Programmed Cell Death 1 Receptor/genetics , Signal Transduction/drug effects
4.
Biomater Sci ; 8(8): 2074-2083, 2020 Apr 21.
Article in English | MEDLINE | ID: mdl-32215398

ABSTRACT

A number of biological tissues have been reported as behaving in an auxetic manner, defined by a negative Poisson's ratio. This describes the deformation of tissue which expands in the axial and the transverse directions simultaneously while under uniaxial tension; and contracts axially and transversely upon uniaxial compression. The discovery of auxetic behaviour within biological tissues has implications for the recreation of the auxetic loading environment within tissue engineering. Tissue engineers strive to recreate the natural properties of biological tissue and in order to recreate the unique loading environment of cells from auxetic tissue, an auxetic scaffold is required. A number of studies have used a variety of auxetic scaffolds within tissue engineering. Investigation into the effect of auxetic micro-environments created by auxetic scaffolds on cellular behaviour has demonstrated an increased cellular proliferation and enhanced differentiation. Here, we discuss studies which have identified auxetic behaviour within biological tissues, and where cells have been cultured within auxetic scaffolds, bringing together current knowledge of the potential use of auxetic materials in tissue engineering applications and biomedical devices.


Subject(s)
Tissue Engineering , Animals , Humans , Tissue Scaffolds
5.
Biomater Sci ; 7(10): 4310-4324, 2019 Sep 24.
Article in English | MEDLINE | ID: mdl-31410428

ABSTRACT

Intestinal stem cells hold great potential in tissue regeneration of the intestine, however, there are key limitations in their culture in vitro. We previously reported a novel synthetic non-biodegradable hydrogel as a 3D culture model for intestinal epithelium using Caco2 and HT29-MTX cells. Here, we investigated the potential of this system as a 3D scaffold for crypts and single intestinal stem cells to support long-term culture and differentiation. Intestinal crypts were extracted from murine small intestines and Lgr5+ stem cells isolated by magnetic activated cell sorting. Crypts and stem cells were suspended within Matrigel or l-pNIPAM for 14 days or suspended within Matrigel for 7 days then released, dissociated, and suspended within, or on l-pNIPAM hydrogel for 28 days. Cellular behaviour and phenotype were determined by histology and immunohistochemistry for stem cell and differentiation markers: Lgr5, E-cadherin MUC2 chromograninA and lysozymes. Isolated crypts and Lgr5+ intestinal stem cells formed enteroids with a central lumen surrounded by multiple crypt-like buds when cultured in Matrigel. In contrast, when crypts and stem cells were directly suspended within, or layered on l-pNIPAM hydrogel under dynamic culture conditions they formed spherical balls of cells, with no central lumen. When enteroids were initially formed in Matrigel from crypts or single Lgr5+ intestinal stem cells and dissociated into small fragments or single cells and transferred to l-pNIPAM hydrogel they formed new larger enteroids with numerous crypt-like buds. These crypt-like buds showed the presence of mucin-producing cells, which resembled goblet cells, scattered throughout their structures. Immunohistochemistry staining also showed the expression of Lgr5 and differentiation markers of all the main intestinal cell types including: enterocytes, goblet cells, enteroendocrine and Paneth cells. This demonstrated that l-pNIPAM hydrogel supported long-term culture of crypts and Lgr5+ stem cells and promoted intestinal cell differentiation.


Subject(s)
Stem Cells/cytology , Tissue Engineering/methods , Animals , Caco-2 Cells , Cell Differentiation/physiology , Cell Line , Humans , Hydrogels , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestine, Small/cytology , Mice , Mice, Inbred BALB C , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism
6.
Oncotarget ; 10(44): 4570-4586, 2019 Jul 16.
Article in English | MEDLINE | ID: mdl-31360305

ABSTRACT

Polyphenols have been shown to sensitize solid tumours to alkylating agents such as cisplatin, and induce apoptosis and/or cell-cycle arrest. Here, we assess the effects of five polyphenols alone and in combination with three alkylating agents: cisplatin, cyclophosphamide and chlorambucil in lymphoid and myeloid leukaemia cells lines, and non-tumour control cells. In lymphoid leukaemia cell lines there was a synergistic reduction in ATP and glutathione levels, an induction of cell cycle arrest, DNA damage and apoptosis when quercetin, apigenin, emodin and rhein were combined with cisplatin and cyclophosphamide; and when apigenin and rhein were combined with chlorambucil. In myeloid leukaemia cells quercetin, apigenin and emodin showed a similar synergistic effect with all alkylating agents; however antagonistic effects were observed with some or all alkylating agents when combined with emodin, rhein and cis-stilbene. All synergistic effects were associated with reduced glutathione levels, DNA damage and apoptosis; whilst during antagonism the reverse effects were observed. The combination of alkylating agents, particularly cisplatin with polyphenols could be promising for the treatment of lymphoid leukaemias, with apigenin showing the greatest effects. Likewise in myeloid cells apigenin also synergised the action of all alkylating agents, suggesting that apigenin may also be beneficial in myeloid leukaemias.

7.
Oncotarget ; 10(37): 3559-3575, 2019 May 28.
Article in English | MEDLINE | ID: mdl-31191826

ABSTRACT

Interleukin 1 (IL-1) is an important mediator of inflammation and tissue damage in inflammatory bowel disease (IBD). The balance between IL-1 and IL-1Ra as a natural inhibitor plays a vital role in a variety of diseases. Here, we investigated whether changes seen during IBD are induced spontaneously in mice lacking a functional IL-1rn gene. Histological staining was performed on the jejunum and ileum of BALB/c IL-1rn+/+ and IL-1rn-/- mice to characterize crypt-villus height, villus width, and number of goblet cells per villus. Pro-inflammatory cytokines, immune cell infiltration and matrix-degrading enzymes, together with the production of intestinal enzymes and the integrity of tight and adherent junction proteins were determined using immunohistochemistry. In the small intestine of BALB/c IL-1rn-/- mice the villus heights were significantly reduced; and in the ileum this was accompanied by a decrease in villi width. There was also an increase in goblet cell number and mucin production compared to wild-type mice. IL-1α and IL-1ß immunopositivity were increased, whilst IL-1R1 expression was decreased in IL-1rn-/- mice. IL-15 and TNFα were also increased in older IL-1rn-/- mice. Increased polymorphonuclear and macrophage infiltration were seen in IL-1rn-/- mice, whilst expression of matrix-degrading enzymes and digestive enzymes were unchanged, except for dipeptidyl peptidase IV which was increased in younger IL-1rn-/- mice compared to wild type mice. The expression of tight and adhesion junctions were also dramatically decreased in IL-1rn-/- mice. In conclusion, IL-1rn-/- mice developed spontaneous abnormalities which displayed features associated with IBD, demonstrating a clear role for IL-1 in IBD.

8.
Sci Rep ; 9(1): 1812, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30755679

ABSTRACT

The in vitro study of the pathogenesis of inflammatory bowel disease (IBD) requires a cell model which closely reflects the characteristics of the in vivo intestinal epithelium. This study aimed to investigate the application of L-pNIPAM hydrogel as a scaffold to develop a long-term 3D co-culture model of Caco-2 and HT29-MTX cells under conditions analogous to inflammation, to determine its potential use in studying IBD. Monocultures and co-cultures were layered on L-pNIPAM hydrogel scaffolds and maintained under dynamic culture conditions for up to 12 weeks. Treatments with IL-1ß, TNFα, and hypoxia for 1 week were used to create an inflammatory environment. Following prolonged culture, the metabolic activity of Caco-2 monoculture and 90% Caco-2/10% HT29-MTX co-cultures on L-pNIPAM hydrogels were increased, and finger-like structures, similar in appearance to villi were observed. Following treatment with IL-1ß, TNFα and hypoxia, ALP and ZO-1 were decreased, MUC2 increased, and MUC5AC remained unchanged. ADAMTS1 was increased in response to hypoxia. Caspase 3 expression was increased in response to TNFα and hypoxic conditions. In conclusion, L-pNIPAM hydrogel supported long-term co-culture within a 3D model. Furthermore, stimulation with factors seen during inflammation recapitulated features seen during IBD.


Subject(s)
Coculture Techniques/methods , Hydrogels/chemistry , Inflammatory Bowel Diseases/metabolism , ADAMTS1 Protein/metabolism , Caco-2 Cells , Caspase 3/metabolism , Cell Hypoxia/drug effects , HT29 Cells , Humans , Interleukin-1beta/pharmacology , Mucin 5AC/metabolism , Mucin-2/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Zonula Occludens-1 Protein/metabolism
9.
Tissue Eng Part B Rev ; 24(2): 98-111, 2018 04.
Article in English | MEDLINE | ID: mdl-28922991

ABSTRACT

In recent years, three-dimensional (3D) cell culture models of the small intestine have gained much attention. These models support cell proliferation, migration, and differentiation and encourage tissue organization which is not possible in two-dimensional (2D) culture systems. Furthermore, the use of a wide variety of cell culture scaffolds and support substrates has revealed considerable differences in cell behavior and tissue organization. These systems have been used in combination with intestinal stem cells, organoid units, or human colonic adenocarcinoma cell lines such as Caco-2 and HT29-MTX to generate a number of in vitro and in vivo models of the intestine. In this study, we review the current 2D and 3D tissue engineering models of the intestine to determine the most effective sources of intestinal cells and current research on support scaffolds capable of inducing the morphological architecture and function of the intestinal mucosa.


Subject(s)
Intestine, Small , Models, Biological , Tissue Engineering/methods , Animals , Caco-2 Cells , Humans , Tissue Engineering/trends
10.
Oncotarget ; 8(62): 104877-104893, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29285220

ABSTRACT

Polyphenols have been previously shown to sensitize leukemia cell lines to topoisomerase inhibitors. Here, we assess the effects of five polyphenols when used alone and in combination with antimetabolites: methotrexate, 6-mercaptopurine and 5-fluorouracil; in lymphoid and myeloid leukemia cells lines, and non-tumor control cells. The effects of combined treatments were investigated on ATP and glutathione levels, cell-cycle progression, DNA damage and apoptosis. Polyphenols antagonized methotrexate and 6-mercaptopurine induced cell-cycle arrest and apoptosis in most leukemia cell lines. This was associated with reduced DNA damage and increased glutathione levels, greater than that seen following individual treatments alone. In contrast, 5-fluorouracil when combined with quercetin, apigenin and rhein caused synergistic decrease in ATP levels, induction of cell-cycle arrest and apoptosis in some leukemia cell lines. However, antagonistic effects were observed when 5-fluorouracil was combined with rhein and cis-stilbene in myeloid cell lines. The effects were dependant on polyphenol type and chemotherapy agent investigated, and cell type treated. Interestingly treatment of non-tumor control cells with polyphenols protected cells from antimetabolite treatments. This suggests that polyphenols modulate the action of antimetabolite agents; more importantly they antagonized methotrexate and 6-mercaptopurine actions, thus suggesting the requirement of polyphenol-exclusion during their use.

11.
Anticancer Agents Med Chem ; 13(10): 1601-13, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23796248

ABSTRACT

BACKGROUND: Mortality rates for leukemia are high despite considerable improvements in treatment. Since polyphenols exert pro-apoptotic effects in solid tumors, our study investigated the effects of polyphenols in haematological malignancies. The effect of eight polyphenols (quercetin, chrysin, apigenin, emodin, aloe-emodin, rhein, cis-stilbene and trans-stilbene) were studied on cell proliferation, cell cycle and apoptosis in four lymphoid and four myeloid leukemic cells lines, together with normal haematopoietic control cells. METHODS: Cellular proliferation was measured by CellTiter-Glo(®) luminescent assay; and cell cycle arrest was assessed using flow cytometry of propidium iodide stained cells. Apoptosis was investigated by caspase-3 activity assay using flow cytometry and apoptotic morphology was confirmed by Hoescht 33342 staining. RESULTS: Emodin, quercetin, and cis-stilbene were the most effective polyphenols at decreasing cell viability (IC50 values of 5-22 µM, 8-33 µM, and 25-85 µM respectively) and inducing apoptosis (AP50 values (the concentration which 50% of cells undergo apoptosis) of 2-27 µM, 19-50 µM, and 8-50 µM respectively). Generally, lymphoid cell lines were more sensitive to polyphenol treatment compared to myeloid cell lines, however the most resistant myeloid (KG-1a and K562) cell lines were still found to respond to emodin and quercetin treatment at low micromolar levels. Non-tumor cells were less sensitive to all polyphenols compared to the leukemia cells. CONCLUSIONS: These findings suggest that polyphenols have anti-tumor activity against leukemia cells with differential effects. Importantly, the differential sensitivity of emodin, quercetin, and cis-stilbene between leukemia and normal cells suggests that polyphenols are potential therapeutic agents for leukemia.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Emodin/pharmacology , Lymphocytes/drug effects , Myeloid Cells/drug effects , Quercetin/pharmacology , Stilbenes/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Emodin/chemistry , Humans , Inhibitory Concentration 50 , Lymphocytes/pathology , Molecular Structure , Myeloid Cells/pathology , Organ Specificity , Quercetin/chemistry , Stereoisomerism , Stilbenes/chemistry , Structure-Activity Relationship
12.
Food Sci Nutr ; 1(2): 196-208, 2013 Mar.
Article in English | MEDLINE | ID: mdl-24804028

ABSTRACT

Pomegranates have shown great promise as anti-cancer agents in a number of cancers including clinical trials in prostate cancer. We have previously shown pomegranate juice (PGJ) induced apoptosis and preferentially alters the cell cycle in leukemia cell lines compared with nontumor control cells. However, the agents responsible have not yet been fully elucidated. Treatment of four leukemia cell lines with five fractions obtained from PGJ by solid phase extraction demonstrated that only the acetonitrile fractions decreased adenosine triphosphate (ATP) levels in all leukemia cell lines. Acetonitrile fractions also significantly activated caspase-3 and induced nuclear morphology characteristic of apoptosis. S phase arrest was induced by acetonitrile fractions which matched S phase arrest seen previously following whole PGJ treatments. The acetonitrile fractions contained higher phenol content than whole PGJ whereas only low levels of phenols were seen in any other fraction. Liquid chromatography mass spectrometry (LC-MS) analysis demonstrated that acetonitrile fractions were enriched in ellagitannins, ellagic acid, and hydroxycinnamic acid derivatives but depleted in anthocyanins. Individual treatments with identified compounds demonstrated that the ellagitannin: punicalagin was the most active and mimicked the responses seen following acetonitrile fraction treatment. Bioactive components within pomegranate were confined to the acetonitrile fraction of PGJ. The enrichment in ellagitannins and hydroxycinnamic acids suggest these may provide the majority of the bioactivities of PGJ. Individual treatments with compounds identified demonstrated that the ellagitannin: punicalagin was the most active agent, highlighting this compound as a key bioactive agent in PGJ.

13.
Nutr Cancer ; 64(1): 100-10, 2012.
Article in English | MEDLINE | ID: mdl-22098126

ABSTRACT

Studies suggest that pomegranates contain bioactive chemicals with potential for treatment and prevention of cancer. Pomegranate juice extracts (PJE) have been shown to inhibit cellular proliferation and tumor growth and induce cell death via apoptosis in a number of cancer cell lines. However, to date, few studies have investigated the potential of PJE in the treatment of leukemia. We investigated the potential effect of PJE on induction of apoptosis and inhibition of cellular proliferation in 8 leukemia cell lines (4 lymphoid and 4 myeloid) and nontumor hematopoietic stem cells (control cells). Apoptosis was assessed by 2 methods: Annexin V-FITC/propidium iodide staining with flow cytometric analysis and 4'-6-diamidino-2-phenylindole (DAPI) morphological assessment. Cell cycle stage was investigated using propidum iodide staining of DNA content and flow cytometric analysis. Live cell counts were also performed using a trypan exclusion assay. PJE significantly induced apoptosis in all cell lines, including nontumor control cells, although lymphoid cells and 2 of the myeloid cell lines were more sensitive. Furthermore, PJE induced cell cycle arrest. These results were confirmed by DAPI analysis and viable cell counts using trypan blue exclusion assay. Our results provide evidence that PJE contain bioactive compounds that could be used in the treatment of leukemia.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Leukemia/drug therapy , Lythraceae/chemistry , Plant Extracts/pharmacology , Annexins/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Death/drug effects , Drug Screening Assays, Antitumor , Fluorescent Dyes/analysis , Hydrogen-Ion Concentration , Indoles/analysis , Leukemia/pathology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...