Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Chin Med ; 18(1): 61, 2023 May 28.
Article in English | MEDLINE | ID: mdl-37246229

ABSTRACT

BACKGROUND: Pancreatic cancer (PAC), a malignancy that is fatal and commonly diagnosed at a late stage. Despite considerable advancements in cancer treatment, the survival rate of PAC remains largely consistent for the past 60 years. The traditional Chinese medicine formula Pulsatilla Decoction (PD) has been clinically used to treat inflammatory diseases for millennia and recently as a supplementary anti-cancer treatment in China. However, the bioactive ingredients and mechanisms underlying its anti-cancer effect remains unclear. METHODS: The composition and quality control of PD were verified through analysis by high performance liquid chromatography. Cell viability was determined using Cell Counting Kit-8 assay. The cell cycle distribution was analyzed through PI staining and flow cytometry analysis, while apoptotic cells were measured by double staining with Annexin V-FITC and PI. We used immunoblotting to examine protein expressions. The in vivo effects of ß-peltatin and podophyllotoxin were evaluated on a subcutaneously-xenografted BxPC-3 cell nude mice model. RESULTS: The current study demonstrated that PD markedly inhibited PAC cell proliferation and triggered their apoptosis. Four herbal PD formula was then disassembled into 15 combinations of herbal ingredients and a cytotoxicity assay showed that the Pulsatillae chinensis exerted the predominant anti-PAC effect. Further investigation indicated that ß-peltatin was potently cytotoxic with IC50 of ~ 2 nM. ß-peltatin initially arrested PAC cells at G2/M phase, followed by apoptosis induction. Animal study confirmed that ß-peltatin significantly suppressed the growth of subcutaneously-implanted BxPC-3 cell xenografts. Importantly, compared to podophyllotoxin that is the parental isomer of ß-peltatin but clinically obsoleted due to its severe toxicity, ß-peltatin exhibited stronger anti-PAC effect and lower toxicity in mice. CONCLUSIONS: Our results demonstrate that Pulsatillae chinensis and particularly its bioactive ingredient ß-peltatin suppress PAC by triggering cell cycle arrest at G2/M phase and apoptosis.

2.
Cancer Lett ; 554: 216011, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36442771

ABSTRACT

Quiescent cancer cells (QCCs), also known as dormant cancer cells, resist and survive chemo- and radiotherapy, resulting in treatment failure and later cancer recurrence when QCCs resume cell cycle progression. However, drugs selectively targeting QCCs are lacking. Saikosaponin A (SSA) derived from Bupleurum DC., is highly potent in eradicating multidrug-resistant prostate QCCs compared with proliferative prostate cancer cells. By further exacerbating the already increased autophagy through inactivation of Akt-mTOR signaling, SSA triggered cell death in QCCs. Contrarily, inhibition of autophagy or activation of Akt signaling pathway prevented SSA-induced cell death. The multicycle of Docetaxel treatments increased the proportion of QCCs, whereas administering SSA at intervals of Docetaxel treatments aggravated cell death in vitro and led to tumor growth arrest and cell death in vivo. In conclusion, SSA is posed as a novel QCCs-eradicating agent by aggravating autophagy in QCCs. In combination with the current therapy, SSA has potential to improve treatment effectiveness and to prevent cancer recurrence.


Subject(s)
Prostatic Neoplasms , Proto-Oncogene Proteins c-akt , Male , Humans , Docetaxel/pharmacology , Docetaxel/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Prostate/pathology , Cell Line, Tumor , Neoplasm Recurrence, Local , Prostatic Neoplasms/pathology , Autophagy , Apoptosis , Cell Proliferation
3.
Discov Oncol ; 13(1): 44, 2022 Jun 07.
Article in English | MEDLINE | ID: mdl-35670862

ABSTRACT

As a natural flavone, apigenin is abundantly present in vegetables, fruits, oregano, tea, chamomile, wheat sprout and is regarded as a major component of the Mediterranean diet. Apigenin is known to inhibit proliferation in different cancer cell lines by inducing G2/M arrest, but it is unclear whether this action is predominantly imposed on G2 or M phases. In this study, we demonstrate that apigenin arrests prostate cancer cells at G2 phase by flow cytometric analysis of prostate cancer cells co-stained for phospho-Histone H3 and DNA. Concurrently, apigenin also reduces the mRNA and protein levels of the key regulators that govern G2-M transition. Further analysis using chromatin immunoprecipitation (ChIP) confirmed the diminished transcriptional activities of the genes coding for these regulators. Unravelling the inhibitory effect of apigenin on G2-M transition in cancer cells provides the mechanistic understanding of its action and supports the potential for apigenin as an anti-cancer agent.

4.
Molecules ; 26(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34885848

ABSTRACT

Phospholipase A2 (PLA2) enzymes were first recognized as an enzyme activity class in 1961. The secreted (sPLA2) enzymes were the first of the five major classes of human PLA2s to be identified and now number nine catalytically-active structurally homologous proteins. The best-studied of these, group IIA sPLA2, has a clear role in the physiological response to infection and minor injury and acts as an amplifier of pathological inflammation. The enzyme has been a target for anti-inflammatory drug development in multiple disorders where chronic inflammation is a driver of pathology since its cloning in 1989. Despite intensive effort, no clinically approved medicines targeting the enzyme activity have yet been developed. This review catalogues the major discoveries in the human group IIA sPLA2 field, focusing on features of enzyme function that may explain this lack of success and discusses future research that may assist in realizing the potential benefit of targeting this enzyme. Functionally-selective inhibitors together with isoform-selective inhibitors are necessary to limit the apparent toxicity of previous drugs. There is also a need to define the relevance of the catalytic function of hGIIA to human inflammatory pathology relative to its recently-discovered catalysis-independent function.


Subject(s)
Group II Phospholipases A2/metabolism , Drug Development , Group II Phospholipases A2/antagonists & inhibitors , Group II Phospholipases A2/pharmacology , Humans , Neoplasms/diagnosis , Neoplasms/enzymology , Prognosis
5.
Theranostics ; 11(19): 9605-9622, 2021.
Article in English | MEDLINE | ID: mdl-34646389

ABSTRACT

Rationale: Recurrent and metastatic cancers often undergo a period of dormancy, which is closely associated with cellular quiescence, a state whereby cells exit the cell cycle and are reversibly arrested in G0 phase. Curative cancer treatment thus requires therapies that either sustain the dormant state of quiescent cancer cells, or preferentially, eliminate them. However, the mechanisms responsible for the survival of quiescent cancer cells remain obscure. Methods: Dual genome-editing was carried out using a CRISPR/Cas9-based system to label endogenous p27 and Ki67 with the green and red fluorescent proteins EGFP and mCherry, respectively, in melanoma cells. Analysis of transcriptomes of isolated EGFP-p27highmCherry-Ki67low quiescent cells was conducted at bulk and single cell levels using RNA-sequencing. The extracellular acidification rate and oxygen consumption rate were measured to define metabolic phenotypes. SiRNA and inducible shRNA knockdown, chromatin immunoprecipitation and luciferase reporter assays were employed to elucidate mechanisms of the metabolic switch in quiescent cells. Results: Dual labelling of endogenous p27 and Ki67 with differentiable fluorescent probes allowed for visualization, isolation, and analysis of viable p27highKi67low quiescent cells. Paradoxically, the proto-oncoprotein c-Myc, which commonly drives malignant cell cycle progression, was expressed at relatively high levels in p27highKi67low quiescent cells and supported their survival through promoting mitochondrial oxidative phosphorylation (OXPHOS). In this context, c-Myc selectively transactivated genes encoding OXPHOS enzymes, including subunits of isocitric dehydrogenase 3 (IDH3), whereas its binding to cell cycle progression gene promoters was decreased in quiescent cells. Silencing of c-Myc or the catalytic subunit of IDH3, IDH3α, preferentially killed quiescent cells, recapitulating the effect of treatment with OXPHOS inhibitors. Conclusion: These results establish a rigorous experimental system for investigating cellular quiescence, uncover the high selectivity of c-Myc in activating OXPHOS genes in quiescent cells, and propose OXPHOS targeting as a potential therapeutic avenue to counter cancer cells in quiescence.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Ki-67 Antigen/metabolism , Melanoma/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cellular Senescence , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Isocitrate Dehydrogenase/metabolism , Neoplasms/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Resting Phase, Cell Cycle , Transcriptome/genetics
6.
Biomed Pharmacother ; 141: 111795, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34098217

ABSTRACT

Cancer recurrence poses a significant challenge. At the cellular level, recurrence takes place as a result of reactivation of dormant cancer cells residing at G0 phase. The aim of the study was to identify compounds that can trap prostate and lung cancer cells in G0 phase from a new Chinese herb recipe, Astringent recipe, consisting of Radix Paeoniae Alba, Agrimonia pilosa Ledeb, Fructus Mume, Fritillaria thunbergii Miq., Ganoderma Lucidum Karst, and Astragalus membranaceus (Fisch.) Bunge. Astringent recipe impeded cell cycle progression in prostate and lung cancer cells by rounding them up at G0 phase by flow cytometric analysis of cancer cells stained with Hoechst 33342 and Pyronin Y, respectively, for DNA and RNA. The anti-cancer efficacy of the recipe was found to be attributable to Agrimonia pilosa Ledeb. Further study established that agrimol B, a polyphenol derived from Agrimonia pilosa Ledeb, contributed to the activity of the herb. The action of agrimol B on the cancer cells was likely derived from its effect on c-MYC, SKP2 and p27 by immunoblotting and immunofluorescence. Oral administration of Agrimonia pilosa Ledeb or agrimol B reduced growth of prostate cancer cell xenograft in animal. In conclusion, Agrimol B can enrich for prostate and lung cancer cells in G0 state and influence key regulators that govern G0 status.


Subject(s)
Agrimonia , Antineoplastic Agents, Phytogenic/pharmacology , Butanones/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , Phenols/pharmacology , Plant Extracts/pharmacology , Tumor Burden/drug effects , A549 Cells , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Butanones/isolation & purification , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/physiology , Dose-Response Relationship, Drug , Ellagic Acid/pharmacology , G1 Phase Cell Cycle Checkpoints/physiology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Phenols/isolation & purification , Plant Extracts/isolation & purification , Tumor Burden/physiology
7.
Cells ; 10(3)2021 03 05.
Article in English | MEDLINE | ID: mdl-33807533

ABSTRACT

Quiescent cancer cells (QCCs) are cancer cells that are reversibly suspended in G0 phase with the ability to re-enter the cell cycle and initiate tumor growth, and, ultimately, cancer recurrence and metastasis. QCCs are also therapeutically challenging due to their resistance to most conventional cancer treatments that selectively act on proliferating cells. Considering the significant impact of QCCs on cancer progression and treatment, better understanding of appropriate experimental models, and the evaluation of QCCs are key questions in the field that have direct influence on potential pharmacological interventions. Here, this review focuses on existing and emerging preclinical models and detection methods for QCCs and discusses their respective features and scope for application. By providing a framework for selecting appropriate experimental models and investigative methods, the identification of the key players that regulate the survival and activation of QCCs and the development of more effective QCC-targeting therapeutic agents may mitigate the consequences of QCCs.


Subject(s)
Neoplasms/pathology , Cell Line, Tumor , Humans
8.
Curr Dev Nutr ; 4(5): nzaa025, 2020 May.
Article in English | MEDLINE | ID: mdl-32391511

ABSTRACT

Citrus fruit and in particular flavonoid compounds from citrus peel have been identified as agents with utility in the treatment of cancer. This review provides a background and overview regarding the compounds found within citrus peel with putative anticancer potential as well as the associated in vitro and in vivo studies. Historical studies have identified a number of cellular processes that can be modulated by citrus peel flavonoids including cell proliferation, cell cycle regulation, apoptosis, metastasis, and angiogenesis. More recently, molecular studies have started to elucidate the underlying cell signaling pathways that are responsible for the flavonoids' mechanism of action. These growing data support further research into the chemopreventative potential of citrus peel extracts, and purified flavonoids in particular. This critical review highlights new research in the field and synthesizes the pathways modulated by flavonoids and other polyphenolic compounds into a generalized schema.

9.
J Ethnopharmacol ; 251: 112526, 2020 Apr 06.
Article in English | MEDLINE | ID: mdl-31893534

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Hedyotis diffusa Willd. (H) and Scutellaria barbata D.Don (S) are ancient anti-cancer Chinese herb medicines. When combined, known as HS, it is one of the most commonly prescribed Chinese Medicines for cancer patients today in China. AIM OF THE STUDY: The prevention of disease progression is a dominant concern for the growing number of men with prostate cancer. The purpose of this work is to evaluate the action and mode of action of Chinese Medicine recipe HS in inhibiting prostate cancer progression in preclinical models. METHODS: Effects of HS were analyzed in prostate cancer cell lines by evaluating proliferation, cell cycle profile, DNA damage and key regulators responsible for G2 to M phase transition. The transcriptional activities of these regulators were determined by RT-PCR and ChIP. The efficacy of HS in vitro was validated in an animal model. RESULTS: HS treatment was observed to reduce DNA content and accumulated prostate cancer cells at the G2/M phase. Immunolabeling for phospho-Histone H3 in association with nocodazole to capture mitotic cells confirmed that HS impeded G2 to M transition. After excluding DNA damage-induced G2 arrest, it was revealed that HS reduced expression of Cyclin B1, CDK1, PLK1 and Aurora A at both protein and mRNA levels, with concomitant reduction of H3K4 tri-methylation at their promoter-regions. Animals that received oral administration of HS with a dosage relevant to clinical application showed reduced tumor volume and weight with a reduction of Cyclin B1, CDK1, PLK1 and Aurora A protein levels. CONCLUSIONS: HS acts by impeding the G2 to M transition of prostate cancer cells. It is likely that the mode of action is transcriptionally suppressing proteins governing mitotic entry, without eliciting significant DNA damage.


Subject(s)
Antineoplastic Agents, Phytogenic , Cell Cycle Proteins/genetics , Cell Cycle/drug effects , Hedyotis , Plant Extracts , Prostatic Neoplasms , Scutellaria , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Medicine, Chinese Traditional , Mice, Nude , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Transcription, Genetic
10.
J Cell Mol Med ; 24(3): 2229-2239, 2020 02.
Article in English | MEDLINE | ID: mdl-31960591

ABSTRACT

Blockade of cell cycle re-entry in quiescent cancer cells is a strategy to prevent cancer progression and recurrence. We investigated the action and mode of action of CPF mixture (Coptis chinensis, Pinellia ternata and Fructus trichosanthis) in impeding a proliferative switch in quiescent lung cancer cells. The results indicated that CPF impeded cell cycle re-entry in quiescent lung cancer cells by reduction of FACT and c-MYC mRNA and protein levels, with concomitant decrease in H3K4 tri-methylation and RNA polymerase II occupancy at FACT and c-MYC promoter regions. Animals implanted with quiescent cancer cells that had been exposed to CPF had reduced tumour volume/weight. Thus, CPF suppresses proliferative switching through transcriptional suppression of FACT and the c-MYC, providing a new insight into therapeutic target and intervention method in impeding cancer recurrence.


Subject(s)
DNA-Binding Proteins/genetics , High Mobility Group Proteins/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-myc/genetics , Transcription, Genetic/drug effects , Transcriptional Elongation Factors/genetics , A549 Cells , Animals , Araceae/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Promoter Regions, Genetic/drug effects , RNA, Messenger/genetics , Ranunculaceae/chemistry , Trichosanthes/chemistry
11.
Biometals ; 32(6): 861-873, 2019 12.
Article in English | MEDLINE | ID: mdl-31583500

ABSTRACT

The intracellular zinc profiles of breast and prostate cancer cells are diametrically opposed, with hyper-accumulation of zinc in breast cancer, and low level in prostate cancer. This phenomenon is poorly understood. This study employs two breast and two prostate cancer cell lines to investigate the role of protein kinase CK2 in regulating zinc homeostasis. CK2 was targeted by its specific inhibitors 4,5,6,7-tetrabromobenzotriazole (TBB) and CX-4945, and by the specific siRNA against each of the three CK2 genes. The effect of zinc exposure after the above CK2 manipulation was observed by MTT [3-(4,5-dimethyliazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide] cell viability assay and confocal microscopy for intracellular zinc level. The results demonstrate that CK2 is involved in regulating zinc homeostasis in breast and prostate cancer cells as both TBB and CX-4945 substantially decreased cell viability upon zinc exposure. siRNA-mediated knockdown of the three CK2 subunits (α, α' and ß) revealed their discrete roles in regulating zinc homeostasis in breast and prostate cancer cells. Knockdown of CK2α' decreased the intracellular zinc level of breast cancer cells and in turn increased the cell viability while the opposite findings were obtained for the prostate cancer cells. Knockdown of CK2ß expression substantially increased the zinc level in breast cancer cell lines whilst decreased the zinc level in prostate cancer cells. Taken together, this study shows that CK2 is involved in zinc homeostasis of breast and prostate cancer cells and opens a new avenue for research on these cancers.


Subject(s)
Antineoplastic Agents/metabolism , Breast Neoplasms/metabolism , Casein Kinase II/metabolism , Homeostasis , Prostatic Neoplasms/metabolism , Zinc Sulfate/metabolism , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Casein Kinase II/antagonists & inhibitors , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Homeostasis/drug effects , Humans , MCF-7 Cells , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Structure-Activity Relationship , Tumor Cells, Cultured , Zinc Sulfate/pharmacology
12.
Nat Commun ; 10(1): 3319, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31346162

ABSTRACT

Chromosome 17q21-ter is commonly gained in neuroblastoma, but it is unclear which gene in the region is important for tumorigenesis. The JMJD6 gene at 17q21-ter activates gene transcription. Here we show that JMJD6 forms protein complexes with N-Myc and BRD4, and is important for E2F2, N-Myc and c-Myc transcription. Knocking down JMJD6 reduces neuroblastoma cell proliferation and survival in vitro and tumor progression in mice, and high levels of JMJD6 expression in human neuroblastoma tissues independently predict poor patient prognosis. In addition, JMJD6 gene is associated with transcriptional super-enhancers. Combination therapy with the CDK7/super-enhancer inhibitor THZ1 and the histone deacetylase inhibitor panobinostat synergistically reduces JMJD6, E2F2, N-Myc, c-Myc expression, induces apoptosis in vitro and leads to neuroblastoma tumor regression in mice, which are significantly reversed by forced JMJD6 over-expression. Our findings therefore identify JMJD6 as a neuroblastoma tumorigenesis factor, and the combination therapy as a treatment strategy.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/metabolism , Neuroblastoma/drug therapy , Receptors, Cell Surface/metabolism , Animals , Apoptosis/drug effects , Carcinogenesis , Cell Proliferation/drug effects , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , Female , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/administration & dosage , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Mice , Mice, Inbred BALB C , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/physiopathology , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics
13.
Food Funct ; 10(6): 3727-3737, 2019 Jun 19.
Article in English | MEDLINE | ID: mdl-31169845

ABSTRACT

The re-entry of quiescent cancer cells to the cell cycle plays a key role in cancer recurrence, which can pose a high risk after primary treatment. Citrus peel extracts (CPEs) contain compounds that can potentially impair tumour growth; however the mechanism of action and effects on cell cycle regulation remain unclear. In this study, the capacity of an ethyl acetate : hexane extract (CPE/hexane) and water extract (CPE/water) to modulate the cell cycle re-entry of quiescent (PC-3 and LNCaP) prostate cancer cells was tested in an in vitro culture system. Cell cycle analysis showed that the quiescent PC-3 and LNCaP cancer cells in the presence of CPE/water were impaired in their ability to enter the S phase where only 2-3% reduction of G0/G1 cells was noted compared to 12-18% reduction of control cells. In contrast, the CPE/hexane did not show any cell cycle inhibition activity in both cell lines. A low DNA synthesis rate and weak apoptosis were observed in quiescent cancer cells treated with CPEs. Hesperidin and narirutin, the predominant flavonoids found in citrus fruits, were not responsible for the observed biological activity, implicating alternative bioactive compounds. Notably, citric acid was identified as one of the compounds present in CPEs that acts as a cell cycle re-entry inhibitor. Citric acid exhibited a higher cell toxicity effect on PC-3 prostate cancer cells than non-cancerous RWPE-1 prostate cells, suggesting specific benefits for cancer treatment. In conclusion, CPE containing citric acid together with various bioactive compounds may be used as a chemopreventive agent for post-therapy cancer patients.


Subject(s)
Citrus/chemistry , Plant Extracts/pharmacology , Prostatic Neoplasms/physiopathology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Disaccharides/isolation & purification , Disaccharides/pharmacology , Flavanones/isolation & purification , Flavanones/pharmacology , Fruit/chemistry , Hesperidin/isolation & purification , Hesperidin/pharmacology , Humans , Male , Prostatic Neoplasms/drug therapy
14.
Int J Cancer ; 145(1): 164-178, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30548853

ABSTRACT

Cancer cell repopulation through cell cycle re-entry by quiescent (G0 ) cell is thought to be an important mechanism behind treatment failure and cancer recurrence. Facilitates Chromatin Transcription (FACT) is involved in DNA repair, replication and transcription by eviction of histones or loosening their contact with DNA. While FACT expression is known to be high in a range of cancers, the biological significance of the aberrant increase is not clear. We found that in prostate and lung cancer cells FACT mRNA and protein levels were low at G0 compared to the proliferating state but replenished upon cell cycle re-entry. Silencing of FACT with Dox-inducible shRNA hindered cell cycle re-entry by G0 cancer cells, which could be rescued by ectopic expression of FACT. An increase in SKP2, c-MYC and PIRH2 and a decrease in p27 protein levels seen upon cell cycle re-entry were prevented or diminished when FACT was silenced. Further, using mVenus-p27K- infected cancer cells to measure p27 degradation capacity, we confirm that inhibition of FACT at release from quiescence suppressed the p27 degradation capacity resulting in an increased mVenus-p27K- signal. In conclusion, FACT plays an important role in promoting the transition from G0 to the proliferative state and can be a potential therapeutic target to prevent prostate and lung cancer from progression and recurrence.


Subject(s)
DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Transcriptional Elongation Factors/metabolism , A549 Cells , Carbazoles/pharmacology , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , High Mobility Group Proteins/antagonists & inhibitors , High Mobility Group Proteins/genetics , Humans , Lung Neoplasms/genetics , Male , PC-3 Cells , Prostatic Neoplasms/genetics , Resting Phase, Cell Cycle/genetics , Transcriptional Elongation Factors/antagonists & inhibitors , Transcriptional Elongation Factors/genetics
15.
Radiother Oncol ; 128(2): 283-300, 2018 08.
Article in English | MEDLINE | ID: mdl-29929859

ABSTRACT

Radiation therapy is used to treat cancer by radiation-induced DNA damage. Despite the best efforts to eliminate cancer, some cancer cells survive irradiation, resulting in cancer progression or recurrence. Alteration in DNA damage repair pathways is common in cancers, resulting in modulation of their response to radiation. This article focuses on the recent findings about molecules and pathways that potentially can be targeted to sensitize prostate cancer cells to ionizing radiation, thereby achieving an improved therapeutic outcome.


Subject(s)
DNA Damage/radiation effects , DNA Repair/radiation effects , Prostatic Neoplasms/radiotherapy , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/radiation effects , Aurora Kinases/radiation effects , Cell Cycle/radiation effects , Checkpoint Kinase 1/radiation effects , Cyclin-Dependent Kinases/radiation effects , Cyclins/radiation effects , HSP90 Heat-Shock Proteins/radiation effects , Histone Deacetylases/radiation effects , Humans , Hyaluronan Receptors/radiation effects , Hypoxia-Inducible Factor 1, alpha Subunit/radiation effects , Male , Mutation/radiation effects , NEDD8 Protein/radiation effects , Neoplasm Recurrence, Local/etiology , Neoplasm Recurrence, Local/radiotherapy , Neoplasm, Residual , Neoplastic Stem Cells/radiation effects , Phosphatidylinositol 3-Kinases/radiation effects , Poly(ADP-ribose) Polymerases/radiation effects , Proto-Oncogene Proteins c-met/radiation effects , Radiation Tolerance , Radiation, Ionizing , Receptors, Androgen/radiation effects , TOR Serine-Threonine Kinases/radiation effects , Zinc Finger Protein GLI1/radiation effects
16.
Sci Rep ; 7(1): 4984, 2017 07 10.
Article in English | MEDLINE | ID: mdl-28694520

ABSTRACT

Lung cancer represents a major cause of cancer-related death worldwide. Although various tactics and anti-tumor drugs have been used to improve curative effects, five-year survival rate of lung cancer patients remains poor. In this study, we investigated the action and underlying mechanisms of our recently optimized Chinese herbal formula Yangyinjiedu (YYJD) against lung cancer. YYJD significantly inhibits the proliferation of lung cancer cell lines (95-D, A549, H460 and H1975) by inducing cell cycle arrest and senescence in a dose-dependent manner. In particular, YYJD induces significant G2/M phase arrest and inhibits the colony formation of lung cancer cells. Moreover, we found that administration of YYJD could inhibit the growth of xenografted lung cancer cells in nude mice without loss in body weight. Our findings suggest that the herbal formula YYJD is a potential anti-tumor agent against lung cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Cell Cycle/drug effects , Lung Neoplasms/drug therapy , Plant Extracts/administration & dosage , A549 Cells , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Senescence , DNA Damage , Dose-Response Relationship, Drug , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Nude , Plant Extracts/pharmacology , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
17.
Oncotarget ; 7(35): 56650-56663, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27494863

ABSTRACT

Hepatocellular carcinoma (HCC) is an aggressive malignancy and the 5-year survival rate of advanced HCC is < 10%. Guttiferone K (GUTK) isolated from the Garcinia genus inhibited HCC cells migration and invasion in vitro and metastasis in vivo without apparent toxicity. Proteomic analysis revealed that actin-binding protein profilin 1 (PFN1) was markedly increased in the presence of GUTK. Over-expression of PFN1 mimicked the effect of GUTK on HCC cell motility and metastasis. The effect of GUTK on cell motility was diminished when PFN1 was over-expressed or silenced. Over-expression of PFN1 or incubation with GUTK decreased F-actin levels and the expression of proteins involved in actin nucleation, branching and polymerization. Moreover, a reduction of PFN1 protein levels was common in advanced human HCC and associated with poor survival rate. In conclusion, GUTK effectively suppresses the motility and metastasis of HCC cells mainly by restoration of aberrantly reduced PFN1 protein expression.


Subject(s)
Benzophenones/pharmacology , Carcinoma, Hepatocellular/metabolism , Garcinia/chemistry , Liver Neoplasms/metabolism , Plant Extracts/chemistry , Profilins/metabolism , Actins/chemistry , Adult , Aged , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Drug Screening Assays, Antitumor , Female , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis/drug therapy , Proteomics , Treatment Outcome
18.
Anticancer Agents Med Chem ; 16(3): 291-8, 2016.
Article in English | MEDLINE | ID: mdl-26419469

ABSTRACT

The continuous activation of the mitogen-activated protein kinase signaling cascade, typified by the BRAFV600E mutation, is one of the key alterations in melanoma. Accordingly, two BRAF inhibitors (BRAFi), vemurafenib and dabrafenib are utilized to treat melanoma and resulted in an excellent clinical outcome. However, the clinical success is not long-lasting, and the BRAFi resistance and disease progression inevitably occurs in nearly all patients. Endoplasmic reticulum stress-induced unfolded protein response and autophagy have emerged as potential pro-survival mechanisms adopted by melanoma cells in response to BRAFi. In this review, we discuss the role of unfolded protein response and autophagy that are implicated in the development of BRAFi-resistant melanoma and the corresponding strategy aiming at overcoming the intractable clinical problem.


Subject(s)
Autophagy , Drug Resistance, Neoplasm , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/drug therapy , Unfolded Protein Response , Cell Line, Tumor , Disease Progression , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/physiology , Humans , Imidazoles/therapeutic use , Indoles/therapeutic use , MAP Kinase Signaling System , Mutation , Oximes/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/therapeutic use , Treatment Outcome , Vemurafenib
19.
Oncotarget ; 6(33): 34458-74, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26416244

ABSTRACT

Cell cycle re-entry of quiescent cancer cells has been proposed to be involved in cancer progression and recurrence. Cytosolic phospholipase A2α (cPLA2α) is an enzyme that hydrolyzes membrane glycerophospholipids to release arachidonic acid and lysophospholipids that are implicated in cancer cell proliferation. The aim of this study was to determine the role of cPLA2α in cell cycle re-entry of quiescent prostate cancer cells. When PC-3 and LNCaP cells were rendered to a quiescent state, the active form of cPLA2α with a phosphorylation at Ser505 was lower compared to their proliferating state. Conversely, the phospho-cPLA2α levels were resurgent during the induction of cell cycle re-entry. Pharmacological inhibition of cPLA2α with Efipladib upon induction of cell cycle re-entry inhibited the re-entry process, as manifested by refrained DNA synthesis, persistent high proportion of cells in G0/G1 and low percentage of cells in S and G2/M phases, together with a stagnant recovery of Ki-67 expression. Simultaneously, Efipladib prohibited the emergence of Skp2 while maintained p27 at a high level in the nuclear compartment during cell cycle re-entry. Inhibition of cPLA2α also prevented an accumulation of cyclin D1/CDK4, cyclin E/CDK2, phospho-pRb, pre-replicative complex proteins CDC6, MCM7, ORC6 and DNA synthesis-related protein PCNA during induction of cell cycle re-entry. Moreover, a pre-treatment of the prostate cancer cells with Efipladib during induction of cell cycle re-entry subsequently compromised their tumorigenic capacity in vivo. Hence, cPLA2α plays an important role in cell cycle re-entry by quiescent prostate cancer cells.


Subject(s)
Cell Cycle/physiology , Cell Proliferation/physiology , Group IV Phospholipases A2/metabolism , Prostatic Neoplasms/pathology , Animals , Benzoates/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Flow Cytometry , Fluorescent Antibody Technique , Heterografts , Humans , Immunoblotting , Immunohistochemistry , Male , Mice , Mice, Nude , Prostatic Neoplasms/enzymology , Sulfonamides/pharmacology
20.
Int J Biochem Cell Biol ; 68: 9-14, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26279144

ABSTRACT

p27(Kip1) is an inhibitor of a broad spectrum of cyclin-dependent kinases (CDKs), and the loss of a single p27(Kip1) allele is thereby sufficient to increase tumor incidence via CDK-mediated cell cycle entry. As such, down-regulation of p27(Kip1) protein levels, in particular nuclear expressed p27(Kip1), is implicated in both disease progression and poor prognosis in a variety of cancers. p27(Kip1) expression is positively regulated by the transcription factor MENIN, and inhibited by oncogenic transcription factors MYC and PIM. However, regulation of p27(Kip1) protein expression and function is predominantly through post-translational modifications that alter both the cellular localization and the extent of E3 ubiquitin ligase-mediated degradation. Phosphorylation of p27(Kip1) at Thr(187) and Ser(10) is a prerequisite for its degradation via the E3 ubiquitin ligases SKP2 (nuclear) and KPC (cytoplasmic), respectively. Additionally, Ser(10) phosphorylated p27(Kip1) is predominantly localized in the cytoplasm due to the nuclear export protein CRM1. Another E3 ubiquitin ligase, PIRH2, degrades p27(Kip1) in both the cytoplasm and nucleus independent of phosphorylation state. As such, inhibition of cell cycle entry and progression in a variety of cancers may be achieved with therapies designed to correct p27(Kip1) localization and/or block its degradation.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Protein Processing, Post-Translational , Transcription, Genetic , Cell Cycle/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Disease Progression , Humans , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...