1
|
Abedin Y, Fife A, Samuels CA, Wright R, Murphy T, Zhang X, Alpert E, Cheung E, Zhao Q, Einstein MH, Douglas NC. Combined Treatment of Uterine Leiomyosarcoma with Gamma Secretase Inhibitor MK-0752 and Chemotherapeutic Agents Decreases Cellular Invasion and Increases Apoptosis. Cancers (Basel) 2024; 16:2184. [PMID: 38927890 PMCID: PMC11201464 DOI: 10.3390/cancers16122184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/23/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Due to limited effective therapeutics for uterine leiomyosarcoma (uLMS), the impact of the gamma secretase inhibitor (GSI) MK-0752 with common chemotherapeutics was explored in uLMS. MTT assays were performed on two human uLMS cell lines, SK-UT-1B and SK-LMS-1, using MK-0752, docetaxel, doxorubicin, and gemcitabine, individually and in combination, to determine cell viability after treatment. Synergistic combinations were used in transwell invasion assays, cell cycle flow cytometry, proliferation assays, and RNA sequencing. In SK-UT-1B, MK-0752 was synergistic with doxorubicin and gemcitabine plus docetaxel. In SK-LMS-1, MK-0752 was synergistic with all individual agents and with the combination of gemcitabine plus docetaxel. MK-0752, gemcitabine, and docetaxel decreased invasion in SK-UT-1B 2.1-fold* and in SK-LMS-1 1.7-fold*. In SK-LMS-1, invasion decreased 1.2-fold* after treatment with MK-0752 and docetaxel and 2.2-fold* after treatment with MK-0752 and doxorubicin. Cell cycle analysis demonstrated increases in the apoptotic sub-G1 population with MK-0752 alone in SK-UT-1B (1.4-fold*) and SK-LMS-1 (2.7-fold**), along with increases with all combinations in both cell lines. The combination treatments had limited effects on proliferation, while MK-0752 alone decreased proliferation in SK-LMS-1 (0.63-fold**). Both MK-0752 alone and in combination altered gene expression and KEGG pathways. In conclusion, the combinations of MK-0752 with either doxorubicin, docetaxel, or gemcitabine plus docetaxel are potential novel therapeutic approaches for uLMS. (* p < 0.05, ** p < 0.01).
Collapse
Affiliation(s)
- Yasmin Abedin
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Alexander Fife
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Cherie-Ann Samuels
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Rasheena Wright
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Trystn Murphy
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Xusheng Zhang
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461, USA;
| | - Emily Alpert
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Emma Cheung
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Qingshi Zhao
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Mark H. Einstein
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
| | - Nataki C. Douglas
- Department of Obstetrics, Gynecology, and Reproductive Health, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA; (A.F.); (C.-A.S.); (R.W.); (T.M.); (E.A.); (E.C.); (Q.Z.); (M.H.E.); (N.C.D.)
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, 185 S Orange Avenue, Newark, NJ 07103, USA
| |
Collapse
|
2
|
Talib WH, Ahmed Jum’AH DA, Attallah ZS, Jallad MS, Al Kury LT, Hadi RW, Mahmod AI. Role of vitamins A, C, D, E in cancer prevention and therapy: therapeutic potentials and mechanisms of action. Front Nutr 2024; 10:1281879. [PMID: 38274206 PMCID: PMC10808607 DOI: 10.3389/fnut.2023.1281879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/09/2023] [Indexed: 01/27/2024] Open
Abstract
Cancer, a leading global cause of mortality, arises from intricate interactions between genetic and environmental factors, fueling uncontrolled cell growth. Amidst existing treatment limitations, vitamins have emerged as promising candidates for cancer prevention and treatment. This review focuses on Vitamins A, C, E, and D because of their protective activity against various types of cancer. They are essential as human metabolic coenzymes. Through a critical exploration of preclinical and clinical studies via PubMed and Google Scholar, the impact of these vitamins on cancer therapy was analyzed, unraveling their complicated mechanisms of action. Interestingly, vitamins impact immune function, antioxidant defense, inflammation, and epigenetic regulation, potentially enhancing outcomes by influencing cell behavior and countering stress and DNA damage. Encouraging clinical trial results have been observed; however, further well-controlled studies are imperative to validate their effectiveness, determine optimal dosages, and formulate comprehensive cancer prevention and treatment strategies. Personalized supplementation strategies, informed by medical expertise, are pivotal for optimal outcomes in both clinical and preclinical contexts. Nevertheless, conclusive evidence regarding the efficacy of vitamins in cancer prevention and treatment is still pending, urging further research and exploration in this compelling area of study.
Collapse
Affiliation(s)
- Wamidh H. Talib
- Faculty of Allied Medical Sciences, Applied Science Private University, Amman, Jordan
| | | | - Zeena Shamil Attallah
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman, Jordan
| | - Mohanned Sami Jallad
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman, Jordan
| | - Lina T. Al Kury
- Department of Health Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi, United Arab Emirates
| | - Rawan Wamidh Hadi
- Faculty of Allied Medical Sciences, Applied Science Private University, Amman, Jordan
| | - Asma Ismail Mahmod
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman, Jordan
| |
Collapse
|
3
|
Rezaie H, Alipanah-Moghadam R, Jeddi F, Clark CCT, Aghamohammadi V, Nemati A. Combined dandelion extract and all-trans retinoic acid induces cytotoxicity in human breast cancer cells. Sci Rep 2023; 13:15074. [PMID: 37700002 PMCID: PMC10497591 DOI: 10.1038/s41598-023-42177-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/06/2023] [Indexed: 09/14/2023] Open
Abstract
Breast cancer is one of the most prevalent and deadly cancers among women worldwide. Recently, natural compounds have been widely used for the treatment of breast cancer. Present study evaluated antiproliferative and anti-metastasis activities of two natural compounds of dandelion and all-trans-retinoic acid (ATRA) in human MCF-7 and MDA-MB231 breast cancer cells. We also evaluated the expression of MMP-2, MMP-9, IL-1β, p53, NM23 and KAI1 genes. Data showed a clear additive cytotoxic effect in concentrations of 40 μM ATRA with 1.5 and 4 mg/ml of dandelion extract in MCF-7 and MDA-MB231 cells, respectively. In both cell lines, compared with the untreated cells, the expression levels of MMP-9 and IL-1β were significantly decreased while p53 and KAI1 expression levels were increased. Besides, MMP-2 and NM23 had different expressions in the two studied cell lines. In conclusion, dandelion/ATRA co-treatment, in addition to having strong cytotoxic effects, has putative effects on the expression of anti-metastatic genes in both breast cancer cells.
Collapse
Affiliation(s)
- Hamed Rezaie
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Reza Alipanah-Moghadam
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Farhad Jeddi
- Department of Genetics and Pathology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Cain C T Clark
- Centre for Intelligent Healthcare, Coventry University, Coventry, CV1 5FB, UK
| | | | - Ali Nemati
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
4
|
Pongjantarasatian S, Nowwarote N, Rotchanakitamnuai V, Srirodjanakul W, Saehun R, Janebodin K, Manokawinchoke J, Fournier BPJ, Osathanon T. A γ-Secretase Inhibitor Attenuates Cell Cycle Progression and Invasion in Human Oral Squamous Cell Carcinoma: An In Vitro Study. Int J Mol Sci 2022; 23:8869. [PMID: 36012128 PMCID: PMC9408752 DOI: 10.3390/ijms23168869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 12/13/2022] Open
Abstract
Notch signaling is associated with many human malignancies, including oral squamous cell carcinoma (OSCC). However, the exact function of Notch signaling in OSCC remains unclear. Here, we investigated the effect of Notch signaling inhibition using a γ-secretase inhibitor (DAPT) on OSCC behaviours in vitro. Bioinformatic analysis of public-available gene expression profiles revealed the dysregulation of the Notch signaling pathway in OSCC compared with normal tissues, indicating the role of Notch signaling in OSCC regulation. RNA sequencing analysis of DAPT-treated human OSCC cells revealed the dysregulation of genes related to cell cycle-related pathways. Blocking Notch signaling significantly inhibited cell proliferation. DAPT-induced G0/G1 cell cycle arrest induced cell apoptosis. Furthermore, cell migration and invasion were also reduced in DAPT-treated cells. These findings indicate that Notch signaling activation participates in OSCC regulation by promoting cell growth, cell cycle progression, cell migration, and invasion. These mechanisms could facilitate OSCC progression. These results imply the potential use of Notch signaling inhibitors as a candidate adjuvant treatment in OSCC patients.
Collapse
Affiliation(s)
- Sarai Pongjantarasatian
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nunthawan Nowwarote
- Department of Oral Biology, Faculty of Dentistry, Universite Paris Cite, 75006 Paris, France
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Molecular Oral Pathophysiology, Universite Paris Cite, Sorbonne Universite, 75006 Paris, France
| | - Varumporn Rotchanakitamnuai
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Watcharee Srirodjanakul
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Ritmongkol Saehun
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kajohnkiart Janebodin
- Department of Anatomy, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand
| | - Jeeranan Manokawinchoke
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Benjamin P. J. Fournier
- Department of Oral Biology, Faculty of Dentistry, Universite Paris Cite, 75006 Paris, France
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Molecular Oral Pathophysiology, Universite Paris Cite, Sorbonne Universite, 75006 Paris, France
| | - Thanaphum Osathanon
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
5
|
Butsri S, Kukongviriyapan V, Senggunprai L, Kongpetch S, Prawan A. All- trans-retinoic acid induces RARB-dependent apoptosis via ROS induction and enhances cisplatin sensitivity by NRF2 downregulation in cholangiocarcinoma cells. Oncol Lett 2022; 23:179. [PMID: 35464301 PMCID: PMC9025595 DOI: 10.3892/ol.2022.13299] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 03/03/2022] [Indexed: 11/27/2022] Open
Abstract
All-trans-retinoic acid (ATRA) has been clinically used to treat acute promyelocytic leukemia and is being studied to treat other types of cancer; however, the therapeutic role and mechanism of ATRA against cholangiocarcinoma (CCA) remain unclear. The present study investigated the cytotoxic effect and underlying mechanisms of ATRA on CCA cell lines. Cell viability was evaluated by sulforhodamine B assay. Intracellular reactive oxygen species (ROS) levels were assessed by dihydroethidium assay. Apoptosis analysis was performed by flow cytometry. The pathways of apoptotic cell death induction were examined using enzymatic caspase activity assay. Proteins associated with apoptosis were evaluated by western blotting. The effects on gene expression were analyzed by reverse transcription-quantitative PCR analysis. ATRA induced a concentration- and time-dependent toxicity in CCA cells. Furthermore, when the cytotoxicity of ATRA against retinoic acid receptor (RAR)-deficient cells was assessed, it was revealed that ATRA cytotoxicity was RARB-dependent. Following ATRA treatment, there was a significant accumulation of cellular ROS and ATRA-induced ROS generation led to an increase in the expression levels of apoptosis-inducing proteins and intrinsic apoptosis. Pre-treatment with ROS scavengers could diminish the apoptotic effect of ATRA, suggesting that ROS and mitochondria may have an essential role in the induction of apoptosis. Furthermore, following ATRA treatment, an increase in cellular ROS content was associated with suppressing nuclear factor erythroid 2-related factor 2 (NFE2L2 or NRF2) and NRF2-downstream active genes. ATRA also suppressed cisplatin-induced NRF2 expression, suggesting that the enhancement of cisplatin cytotoxicity by ATRA may be associated with the downregulation of NRF2 signaling. In conclusion, the results of the present study demonstrated that ATRA could be repurposed as an alternative drug for CCA therapy.
Collapse
Affiliation(s)
- Siriwoot Butsri
- Department of Pharmacology, Faculty of Medicine, Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Veerapol Kukongviriyapan
- Department of Pharmacology, Faculty of Medicine, Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Laddawan Senggunprai
- Department of Pharmacology, Faculty of Medicine, Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sarinya Kongpetch
- Department of Pharmacology, Faculty of Medicine, Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Auemduan Prawan
- Department of Pharmacology, Faculty of Medicine, Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
6
|
Increased expression of Profilin potentiates chemotherapeutic agent-mediated tumour regression. Br J Cancer 2022; 126:1410-1420. [PMID: 35022526 PMCID: PMC9091232 DOI: 10.1038/s41416-021-01683-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 12/10/2021] [Accepted: 12/21/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Targeted cancer therapy is an alternative to standard chemotherapy for a better prognosis. Although its incompetency for triple-negative breast cancer (TNBC), treatment still relies on classical chemotherapy. Increasing evidence suggest that chemotherapeutic drug-induced toxic effect could be minimised by combinatorial therapy. Profilin's familiar anti-tumorigenic activity can be utilised in combination with the drug to improve efficacy, which could be promising therapeutics to treat TNBC. METHODS All-trans retinoic acid (ATRA) in combination with vinblastine was tested on human MDA MB-231 cell line (MB-231) (in vitro) and MB-231 borne breast cancer in nude mice (in vivo). Effects of combination treatment on tumour growth inhibition and apoptosis were examined by tumour volume, histology and PARP cleavage. ATRA-induced transcriptional regulation of profilin had been evaluated by gel-shift and reporter gene assays. Profilin's role in ATRA-induced vinblastine efficacy was validated in profilin-stable and profilin-silenced cells. RESULTS ATRA binds with RAR/RXR to increase the profilin expression that potentiated cell death by chemotherapeutics. ATRA priming led to vinblastine-mediated potentiation of G2-M phase cell cycle arrest in MB-231 cells and regression of breast cancer in xenograft mice at very low concentration without any adverse effects. Moreover, increased p53 and PTEN but downregulated p65 in the tumour tissues further supported the involvement of profilin for tumour regression. CONCLUSIONS Vinblastine at very low concentration (20 times lesser than the recommended dose for breast cancer therapeutic) significantly regress tumour growth in ATRA-primed mice without any toxic effects suggesting potential combinatorial therapeutics for TNBC.
Collapse
|
7
|
Acquisition of paclitaxel resistance modulates the biological traits of gastric cancer AGS cells and facilitates epithelial to mesenchymal transition and angiogenesis. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:515-533. [PMID: 35122114 DOI: 10.1007/s00210-022-02217-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022]
Abstract
PURPOSE This study aims to develop a paclitaxel (PTX)-resistant gastric cancer AGS cells (AGS-R) and evaluate the mechanisms of drug resistance. METHODS AGS cells were successively treated with increasing PTX concentrations. Cross-resistance of established AGS-R, the molecular patterns of cell survival, evasion of apoptosis, epithelial-mesenchymal transition (EMT), and the angiogenic potential were evaluated. RESULTS AGS-R was induced within six months of PTX exposure. Extension of the treatment resulted in PTX-resistance beyond clinical levels. The established AGS-R showed resistance to vincristine and doxorubicin but not cisplatin. Upon induction of resistance, the expressions of MDR-1 (P < 0.001) and MRP-1 (P < 0.01) genes and proteins significantly increased. AGS-R cells had elevated levels of BCL-2, pro-CASP3, cleaved-NOTCH1, HES1, HEY1, NF-κB, PI3K, p-AKT, HIF-1α, Cyclin A, and B1 as compared with parental cells (at least P < 0.01). The protein levels of BAX, CASP3, P53, and P21 (at least P < 0.01) as well as intracellular ROS (P < 0.001) were reduced in AGS-R. A relative arrest at the G2/M phase (15.8 ± 0.75 vs. 26.7 ± 1.67) of the cell cycle and enrichment of AGS-R cells for CD44 marker (9 ± 0.6 vs. 1 ± 0.8) (P < 0.001) were detected by flow cytometry. While the E-cadherin expression was reduced (P < 0.001), the protein levels of Vimentin, N-cadherin, SLUG, and SNAIL were increased (at least P < 0.05). The angiogenic activity and release of VEGF and MMP2/9 were increased in AGS-R cells relative to the AGS line (P < 0.001). CONCLUSION AGS-R cells could bypass chemotherapy stress by expressing the genes coding for efflux pumps and altering some key signaling in favor of survival, EMT, and angiogenesis.
Collapse
|
8
|
Jin Y, Teh SS, Lau HLN, Xiao J, Mah SH. Retinoids as anti-cancer agents and their mechanisms of action. Am J Cancer Res 2022; 12:938-960. [PMID: 35411232 PMCID: PMC8984900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/02/2022] [Indexed: 06/14/2023] Open
Abstract
Retinoids (vitamin A) have been reported extensively for anti-cancer properties due to their high receptor-binding affinities and gene regulation abilities. However, the anti-cancer potential of retinoids has not been reviewed in recent years. Thus, this review focused on the anti-cancer effects of retinoids and their synergistic effects with other drugs, together with their mechanisms of action in different types of cancers reported in the past five years. The retinoids were well studied in breast cancer, melanoma, and colorectal cancer. Synthetic retinoids have shown higher selectivity, stronger effectiveness, and lower toxicity than endogenous retinoids. Interestingly, the combination treatment of endogenous retinoids with chemotherapy drugs showed enhanced anti-cancer effects. The mechanisms of action reported for retinoids mainly involved the RAR/RXR signaling pathway. However, limited clinical studies were conducted in recent years. Thus, retinoids which are highly potential anti-cancer agents are worth further study in clinical, especially as a combination therapy with chemotherapy drugs.
Collapse
Affiliation(s)
- Ying Jin
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor’s University (Lakeside Campus)Subang Jaya, Selangor, Malaysia
| | - Soek Sin Teh
- Energy and Environment Unit, Engineering and Processing Division, Malaysian Palm Oil BoardKajang, Selangor, Malaysia
| | - Harrison Lik Nang Lau
- Energy and Environment Unit, Engineering and Processing Division, Malaysian Palm Oil BoardKajang, Selangor, Malaysia
| | - Jianbo Xiao
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense CampusOurense, Spain
| | - Siau Hui Mah
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor’s University (Lakeside Campus)Subang Jaya, Selangor, Malaysia
- Centre for Drug Discovery and Molecular Pharmacology, Faculty of Health and Medical Sciences, Taylor’s University (Lakeside Campus)Subang Jaya, Selangor, Malaysia
| |
Collapse
|
9
|
Xu X, Wu Y, Li H, Xie J, Cao D, Huang X. Notch pathway inhibitor DAPT accelerates in vitro proliferation and adipogenesis in infantile hemangioma stem cells. Oncol Lett 2021; 22:854. [PMID: 34777588 PMCID: PMC8581475 DOI: 10.3892/ol.2021.13115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 06/03/2021] [Indexed: 11/06/2022] Open
Abstract
The Notch signaling pathway is crucial in both adipogenesis and tumor development. It serves a vital role in the development and stability of blood vessels and may be involved in the proliferative phase of infantile hemangiomas, which express various related receptors. Therefore, it was hypothesized that the Notch signaling pathway inhibitor N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT), a γ-secretase inhibitor, might help accelerate the regression of infantile hemangiomas. The present in vitro study evaluated whether inhibition of the Notch signaling pathway using DAPT could alter adipogenesis in hemangioma stem cells (HemSCs) derived from infantile hemangioma (IH) specimens. A total of 20 infants (age, ≤6 months) with hemangiomas who had not yet received any treatment were selected, and their discarded hemangioma tissues were obtained. HemSCs were isolated from the fresh, sterile IH specimens and treated with DAPT. Reverse transcription-quantitative PCR and western blotting were used to demonstrate the inhibition of the Notch signaling pathway by DAPT. A proliferation assay (Cell Counting Kit-8), oil red O staining, flow cytometry and a transwell assay were used to detect proliferation, adipogenesis, apoptosis and migration of HemSCs. Treatment with DAPT upregulated the expression levels of CCAAT/enhancer-binding protein (C/EBP) α, C/EBPβ, peroxisome proliferator-activated receptor-γ, adiponectin and insulin-like growth factor 1, and promoted the proliferation, apoptosis, migration and lipid accumulation in HemSCs in vitro. Targeting the Notch signaling pathway using DAPT may potentially accelerate the regression of infantile hemangiomas.
Collapse
Affiliation(s)
- Xing Xu
- Department of Plastic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Yao Wu
- Department of Plastic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Honghong Li
- Department of Plastic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Juan Xie
- Department of Plastic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Dongsheng Cao
- Department of Plastic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230000, P.R. China
| | - Xueying Huang
- Department of Anatomy, Anhui Medical University, Hefei, Anhui 230000, P.R. China
| |
Collapse
|
10
|
Kim B, Seo Y, Kwon JH, Shin Y, Kim S, Park SJ, Park JJ, Cheon JH, Kim WH, Il Kim T. IL-6 and IL-8, secreted by myofibroblasts in the tumor microenvironment, activate HES1 to expand the cancer stem cell population in early colorectal tumor. Mol Carcinog 2021; 60:188-200. [PMID: 33544929 DOI: 10.1002/mc.23283] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/15/2021] [Accepted: 01/17/2021] [Indexed: 12/24/2022]
Abstract
Interaction between a tumor and its microenvironment is important for tumor initiation and progression. Cancer stem cells (CSCs) within the tumor interact with a microenvironmental niche that controls their maintenance and differentiation. We investigated the CSC-promoting effect of factors released from myofibroblasts into the microenvironment of early colorectal cancer tumors and its molecular mechanism. By messenger RNA microarray analysis, expression of HES1, a Notch signaling target, significantly increased in Caco-2 cells cocultured with 18Co cells (pericryptal myofibroblasts), compared to its expression in Caco-2 cells cultured alone. Caco-2 cells cultured in 18Co-conditioned media (CM) showed a significant increase in CD133+CD44+ cells and HES1 expression compared to that in Caco-2 cells cultured in regular media. Significant amounts of interleukin-6 (IL-6) and IL-8 were detected in 18Co-CM compared to levels in regular media. The 18Co-CM-induced increase in CD133+CD44+ cells was attenuated by IL-6- and IL-8-neutralizing antibodies. Furthermore, these neutralizing antibodies and inhibitors of STAT3 and gamma-secretase reduced the expression of HES1 induced in Caco-2 cells cultured in 18Co-CM. Immunohistochemical analysis of human tissues revealed that IL-6, IL-8, and HES1 expression increased from normal to adenoma, and from adenoma to cancer tissues. In addition, IL-6 and HES1 expression was positively correlated in early colorectal cancer tissues. In conclusion, the increase of CSCs by myofibroblasts could be mediated by IL-6/IL-8-induced HES1 activation in the tumor microenvironment. Based on these data, the IL-6/IL-8-mediated Notch/HES1 and STAT3 pathway, through which CSCs interact with their microenvironment, might be a potential target for the prevention and treatment of colorectal tumors.
Collapse
Affiliation(s)
- Bun Kim
- Department of Medicine, The Graduate School, Yonsei University College of Medicine, Seoul, Korea.,Division of Translational Science, Center for Colon Cancer, Center for Cancer Prevention and Detection, National Cancer Center, Goyang, Korea.,Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Yoojeong Seo
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Ji-Hee Kwon
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Youmi Shin
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
| | - Suhyun Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Soo Jung Park
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Jun Park
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea.,Yonsei Cancer Prevention Center, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Hee Cheon
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Won Ho Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Tae Il Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea.,Yonsei Cancer Prevention Center, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
11
|
Zhang C, Zhang J, Wang J, Yan Y, Zhang C. Alpha-fetoprotein accelerates the progression of hepatocellular carcinoma by promoting Bcl-2 gene expression through an RA-RAR signalling pathway. J Cell Mol Med 2020; 24:13804-13812. [PMID: 33090723 PMCID: PMC7753843 DOI: 10.1111/jcmm.15962] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/19/2020] [Accepted: 08/31/2020] [Indexed: 12/25/2022] Open
Abstract
Previous studies have found that alpha-fetoprotein (AFP) can promote the proliferation of hepatoma cells and accelerate the progression of hepatocellular carcinoma (HCC). However, the exact mechanism of action remains unclear. Recent bioinformatics studies have predicted the possible interaction between AFP and retinoic acid receptors (RARs). Thus, the purpose of this study was to investigate the molecular mechanism through which AFP promotes tumour cell proliferation by interfering with the RA-RAR signal pathway. Our data indicated that AFP could significantly promote the proliferation and weaken ATRA-induced apoptosis of hepatoma cells. Besides, cytoplasmic AFP interacts with RAR, disrupting its entrance into the nucleus, which in turn affects the expression of the Bcl-2 gene. In addition, knockdown of AFP in HepG2 cells was synchronously associated with an incremental increase of RAR binding to DNA, as well as down-regulation of Bcl-2; the opposite effect was observed in AFP gene-transfected HLE cells. Moreover, a similar effect of AFP was detected in tumour tissues with high serum AFP, but not in adjacent non-cancerous liver tissues, or HCC tissues with low serum AFP levels. These results indicate that AFP acts as signalling molecule and prevents RAR from entering into the nucleus by interacting with RAR, thereby promoting the expression of Bcl-2. Our data reveal a novel mechanism through which AFP regulates Bcl-2 expression and further suggest that AFP may be used as a novel target for treating HCC.
Collapse
Affiliation(s)
- Chao Zhang
- National Center for Clinical LaboratoriesNational Center of GerontologyBeijing HospitalBeijingChina
- Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijing HospitalBeijingChina
| | - Jiangtao Zhang
- National Center for Clinical LaboratoriesNational Center of GerontologyBeijing HospitalBeijingChina
- Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijing HospitalBeijingChina
| | - Jing Wang
- National Center for Clinical LaboratoriesNational Center of GerontologyBeijing HospitalBeijingChina
- Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijing HospitalBeijingChina
| | - Ying Yan
- National Center for Clinical LaboratoriesNational Center of GerontologyBeijing HospitalBeijingChina
- Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijing HospitalBeijingChina
| | - Chuanbao Zhang
- National Center for Clinical LaboratoriesNational Center of GerontologyBeijing HospitalBeijingChina
- Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijing HospitalBeijingChina
| |
Collapse
|
12
|
The Neuroprotective Effect of Mesna on Cisplatin-Induced Neurotoxicity: Behavioral, Electrophysiological, and Molecular Studies. Neurotox Res 2020; 39:826-840. [PMID: 33216283 DOI: 10.1007/s12640-020-00315-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 11/07/2020] [Accepted: 11/15/2020] [Indexed: 02/07/2023]
Abstract
Peripheral neuropathy and cognitive impairments following cisplatin administration may interfere with the clinical usage of the drug. Mesna is a chemoprotective agent with anti-inflammatory and anti-oxidant effects. Our study aimed to investigate the protective effects of mesna against cisplatin-induced neurotoxicity. Neurotoxicity was induced by the administration of 2.5 mg/kg cisplatin twice a week for four consecutive weeks in male Wistar rats. The neuroprotective effect of mesna (150 mg/kg/day) was evaluated through behavioral, electrophysiological, and molecular studies. Cisplatin treatment caused passive avoidance memory impairment, increased anxiety-like behaviors, altered thermal sensitivity, and decreased muscle strength in a grip strength test. Our electrophysiological studies indicated that administration of cisplatin induced peripheral sensory neuropathy and decreased the amplitudes of the compound action potential of sensory nerves. Cisplatin administration increased MDA and 4-HNE levels and decreased anti-oxidant (SOD and GPx) enzymes. Proinflammatory cytokines (IL-1β and TNF-α) and metalloproteinase-2 and 9 (MMP-2/9) were increased by cisplatin treatment. Morphological alterations were observed in the dorsal root ganglion (DRG) of cisplatin-treated rats. Cognitive impairments, anxiety, muscle strength, and thermal sensitivity changes induced by cisplatin were improved with mesna treatment. The reduced conduction velocity in sensory nerves was recovered in the cisplatin + mesna group. Mesna partially alleviated redox imbalance, reduced the proinflammatory cytokines, and MMP-2/9 levels. Mesna administration also relieved the morphological changes in DRG of cisplatin-treated rats. In conclusion, our results revealed that mesna can alleviate cisplatin-induced central and peripheral nervous system toxicity. These results support the concept that chemotherapy-induced neuropathy can be partially inhibited via mesna.
Collapse
|
13
|
Xiao N, Hu Y, Juan L. Comprehensive Analysis of Differentially Expressed lncRNAs in Gastric Cancer. Front Cell Dev Biol 2020; 8:557. [PMID: 32695786 PMCID: PMC7338654 DOI: 10.3389/fcell.2020.00557] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/11/2020] [Indexed: 01/26/2023] Open
Abstract
Gastric cancer (GC) is the fourth most common malignant tumor. The mechanism underlying GC occurrence and development remains unclear. Previous studies have indicated that long non-coding RNAs (lncRNAs) are significantly associated with gastric cancer, but a systematic understanding of the role of lncRNAs in gastric cancer is lacking. In recent years, with the development of next-generation sequencing technology, tens of thousands of lncRNAs have been discovered. However, a large number of unannotated lncRNAs remain unidentified in different tissues, including potential gastric cancer-related lncRNAs. In this study, RNA sequencing (RNA-seq) data from 16 samples of eight gastric cancer patients were obtained and analyzed. A total of 1,854 previously unannotated lncRNAs were identified by ab initio assembly, and 520 differentially expressed lncRNAs were validated in the TCGA expression dataset. Methylation and copy number variation (CNV) array data from the same sample were integrated in the analysis. Changes in DNA methylation levels and CNVs may be responsible for the differential expression of 91 lncRNAs. Differentially expressed lncRNAs were enriched in coexpressed clusters of genes related to functions such as cell signaling, cell cycle, immune response, metabolic processes, angiogenesis, and regulation of retinoic acid (RA) receptors. Finally, a differentially expressed lncRNA, AC004510.3, was identified as a potential biomarker for the prediction of the overall survival of gastric cancer patients.
Collapse
Affiliation(s)
- Nan Xiao
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, China.,School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Yang Hu
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, China
| | - Liran Juan
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
14
|
Liu Y, Liu Q, Chen S, Liu Y, Huang Y, Chen P, Li X, Gao G, Xu K, Fan S, Zeng Z, Xiong W, Tan M, Li G, Zhang W. APLNR is involved in ATRA-induced growth inhibition of nasopharyngeal carcinoma and may suppress EMT through PI3K-Akt-mTOR signaling. FASEB J 2019; 33:11959-11972. [PMID: 31408612 DOI: 10.1096/fj.201802416rr] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 07/16/2019] [Indexed: 01/03/2025]
Abstract
The apelin receptor (APLNR) is a GPCR involved in many pathophysiological processes; however, the correlation between APLNR expression and nasopharyngeal carcinoma (NPC) has not been reported. In this study, we used cDNA microarray data to determine APLNR expression levels in NPC tissues. We found that APLNR expression was reduced in NPC tissues compared with noncancerous nasopharyngeal epithelial tissues. Subsequently, a large-scale sample of 1015 tissues was used to validate this discovery and explore the relationship between APLNR expression and prognosis of NPC. Expression levels of APLNR in NPC tissues were indeed down-regulated. Furthermore, positive expression of APLNR in NPC predicted a better prognosis (disease-free survival: P = 0.001; overall survival: P < 0.001). Moreover, ingenuity pathway analysis revealed that an indirect interaction existed between APLNR and retinoic acid (RA) in the cancer regulatory network. Consistently, after treatment with all-trans-RA (ATRA), we found that APLNR was significantly up-regulated in NPC cell lines (5-8F and HNE1), and proliferation of NPC cells was inhibited. Cell cycle arrest occurred in the G0/G1 phase. In contrast, knockdown of APLNR diminished ATRA-induced growth inhibition of NPC cells. In addition, we surprisingly found that APLNR also played an important role in migration and invasion of NPC. Wound-healing and Transwell assays revealed that APLNR overexpression led to reduced migratory and invasive properties in 2 NPC cell lines. Western blot results revealed that hallmarks of epithelial-mesenchymal transition (EMT) were altered as well, suggesting that APLNR was capable of inhibiting EMT in NPC cells. Our study further demonstrated that low expression of APLNR promoted EMT in NPC cells by activating the PI3K-protein kinase B-mammalian target of rapamycin signaling pathway. Taken together, our data suggest that APLNR could potentially predict prognosis for patients with NPC and inhibit proliferation, migration, invasion, and EMT in nasopharyngeal cancer cells.-Liu, Y., Liu, Q., Chen, S., Liu, Y., Huang, Y., Chen, P., Li, X., Gao, G., Xu, K., Fan, S., Zeng, Z., Xiong, W., Tan, M., Li, G., Zhang, W. APLNR is involved in ATRA-induced growth inhibition of nasopharyngeal carcinoma and may suppress EMT through PI3K-Akt-mTOR signaling.
Collapse
Affiliation(s)
- Yi Liu
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qingluan Liu
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Shumin Chen
- Department of Hematology, Peking University People's Hospital, Beijing, China
| | - Yijun Liu
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yumei Huang
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Pan Chen
- Hunan Cancer Hospital-The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xiayu Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Ge Gao
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Keqian Xu
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhaoyang Zeng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Wei Xiong
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Ming Tan
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
- Mitchell Cancer Institute, USA Health-University of South Alabama, Mobile, Alabama, USA
| | - Guiyuan Li
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Wenling Zhang
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
15
|
Mahmoudinia S, Niapour A, Ghasemi Hamidabadi H, Mazani M. 2,4-D causes oxidative stress induction and apoptosis in human dental pulp stem cells (hDPSCs). ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:26170-26183. [PMID: 31280441 DOI: 10.1007/s11356-019-05837-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 06/24/2019] [Indexed: 06/09/2023]
Abstract
2,4-Dicholorophenoxy acetic acid (2,4-D) is a worldwide used hormone herbicide. Human dental pulp stem cells (hDPSCs) as a potential source of mesenchymal stem cells provide a confident model system for the assessments of chemicals in vitro. The main objective of this study was to examine the biological effects and damages attributed to 2,4-D on hDPSCs. hDPSCs were isolated from third molar pulp tissues and their mesenchymal identity were evaluated. Then, hDPSCs were treated with increasing concentrations of 2,4-D (0.1 μM-10 mM). Cell viability assay and cumulative cell counting were carried out to address 2,4-D effects on biological parameters of hDPSCs. Cell cycle distribution, ROS level and ALP activity were measured before and after treatment. AO/EB staining and caspase 3/7 activity were investigated to detect the possible mechanisms of cell death. Flow-cytometric immunophenotyping and differentiation data confirmed the mesenchymal identity of cultivated hDPSCs. 2,4-D treatment caused a hormetic response in the viability and growth rate of hDPSCs. G0/G1 cell cycle arrest, enhanced ROS level, and reduced ALP activity were detected in hDPSCs treated with EC50 dose of 2,4-D. AO/EB staining showed a higher percentage of alive cells in lower concentrations of the herbicide. The increment in 2,4-D dose and the number of early and late apoptotic cells were increased. DAPI staining and caspase 3/7 assay validated the induction of apoptosis. 2,4-D concentrations up to 100 μM did not affect hDPSCs viability and proliferation. The intense cellular oxidative stress and apoptosis were observed at higher concentration.
Collapse
Affiliation(s)
- Samira Mahmoudinia
- Department of Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Niapour
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Hatef Ghasemi Hamidabadi
- Immunogenetic Research Center, Department of Anatomy and Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Mazani
- Department of Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
16
|
Niapour A, Ghasemi Hamidabadi H, Niapour N, Mohammadi P, Sharifi Pasandi M, Malekzadeh V. Pharmacological Notch pathway inhibition leads to cell cycle arrest and stimulates ascl1 and neurogenin2 genes expression in dental pulp stem cells-derived neurospheres. Biotechnol Lett 2019; 41:873-887. [PMID: 31073804 DOI: 10.1007/s10529-019-02687-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 05/05/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Human dental pulp-derived stem cells (hDPSCs) are becoming an attractive source for cell-based neurorestorative therapies. As such, it is important to understand the molecular mechanisms that regulate the differentiation of hDPSCs toward the neuronal fate. Notch signaling plays key roles in neural stem/progenitor cells (NS/PCs) maintenance and prevention of their differentiation. The aim of this study was to address the effects of Notch signaling inhibition on neurosphere formation of hDPSCs and neuronal differentiation of hDPSCs-neurospheres. RESULTS hDPSCs were isolated from third molar teeth. The cultivated hDPSCs highly expressed CD90 and CD44 and minimally presented CD34 and CD45 surface markers. The osteo/adipogenic differentiation of hDPSCs was documented. hDPSCs were cultured in neural induction medium and N-[N-(3,5-difluorophenacetyl-L-alanyl)]-Sphenylglycine t-butyl ester (DAPT) was applied to impede Notch signaling during transformation into spheres or on the formed neurospheres. Our results showed that the size and number of neurospheres decreased and the expression profile of nestin, sox1 and pax6 genes reduced provided DAPT. Treatment of the formed neurospheres with DAPT resulted in the cleaved Notch1 reduction, G0/G1 arrest and a decline in L-lactate production. DAPT significantly reduced hes1 and hey1 genes, while ascl1 and neurogenin2 expressions augmented. The number of MAP2 positive cells improved in the DAPT-treated group. CONCLUSIONS Our findings demonstrated the Notch activity in hDPSCs-neurospheres. DAPT treatment positively regulated proneural genes expression and increased neuronal-like differentiation.
Collapse
Affiliation(s)
- Ali Niapour
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Hatef Ghasemi Hamidabadi
- Department of Anatomy and Cell Biology, Immunogenetic Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Department of Anatomy & Cell Biology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nazila Niapour
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Perham Mohammadi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Marzieh Sharifi Pasandi
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Vadoud Malekzadeh
- Research Laboratory for Embryology and Stem Cells, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
17
|
Sun J, Mu H, Yu J, Li L, Yan H, Li G, Tan H, Yang N, Yang X, Yi L. Diallyl disulfide down-regulates calreticulin and promotes C/EBPα expression in differentiation of human leukaemia cells. J Cell Mol Med 2018; 23:194-204. [PMID: 30394654 PMCID: PMC6307788 DOI: 10.1111/jcmm.13904] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 08/20/2018] [Indexed: 12/31/2022] Open
Abstract
Diallyl disulfide (DADS), the main active component of the cancer fighting allyl sulfides found in garlic, has shown potential as a therapeutic agent in various cancers. Previous studies showed DADS induction of HL-60 cell differentiation involves down-regulation of calreticulin (CRT). Here, we investigated the mechanism of DADS-induced differentiation of human leukaemia cells and the potential involvement of CRT and CCAAT enhancer binding protein-α (C/EBPα). We explored the expression of CRT and C/EBPα in clinical samples (20 healthy people and 19 acute myeloid leukaemia patients) and found that CRT and C/EBPα expressions were inversely correlated. DADS induction of differentiation of HL-60 cells resulted in down-regulated CRT expression and elevated C/EBPα expression. In severe combined immunodeficiency mice injected with HL-60 cells, DADS inhibited the growth of tumour tissue and decreased CRT levels and increased C/EBPα in vivo. We also found that DADS-mediated down-regulation of CRT and up-regulation of C/EBPα involved enhancement of reactive oxidative species. RNA immunoprecipitation revealed that CRT bound C/EBPα mRNA, indicating its regulation of C/EBPα mRNA degradation by binding the UG-rich element in the 3' untranslated region of C/EBPα. In conclusion, the present study demonstrates the C/EBPα expression was correlated with CRT expression in vitro and in vivo and the molecular mechanism of DADS-induced leukaemic cell differentiation.
Collapse
Affiliation(s)
- Jing Sun
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China.,Biology Research Institute, College of Pharmacy and Biological Sciences, University of South China, Hengyang, Hunan, China
| | - Hongxiang Mu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China.,Biology Research Institute, College of Pharmacy and Biological Sciences, University of South China, Hengyang, Hunan, China
| | - Jia Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China.,Biology Research Institute, College of Pharmacy and Biological Sciences, University of South China, Hengyang, Hunan, China
| | - Linwei Li
- Department of Laboratory, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Hongxia Yan
- Department of Laboratory, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Guoqing Li
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China.,Biology Research Institute, College of Pharmacy and Biological Sciences, University of South China, Hengyang, Hunan, China
| | - Hui Tan
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China.,Biology Research Institute, College of Pharmacy and Biological Sciences, University of South China, Hengyang, Hunan, China
| | - Nanyang Yang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China.,Biology Research Institute, College of Pharmacy and Biological Sciences, University of South China, Hengyang, Hunan, China
| | - Xiaoyan Yang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China.,Biology Research Institute, College of Pharmacy and Biological Sciences, University of South China, Hengyang, Hunan, China
| | - Lan Yi
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan, China.,Biology Research Institute, College of Pharmacy and Biological Sciences, University of South China, Hengyang, Hunan, China
| |
Collapse
|
18
|
Bouriez D, Giraud J, Gronnier C, Varon C. Efficiency of All-Trans Retinoic Acid on Gastric Cancer: A Narrative Literature Review. Int J Mol Sci 2018; 19:ijms19113388. [PMID: 30380687 PMCID: PMC6275086 DOI: 10.3390/ijms19113388] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer (GC) is the third leading cause of cancer-related death worldwide with a five-year survival rate of around 25%, and 4% when diagnosed at a metastatic stage. Cancer stem cells (CSC) have recently been characterized as being responsible for resistance to radio/chemotherapies and metastasis formation, opening up perspectives for new targeted therapies. Those CSCs express biomarkers such as cluster of differentiation 44 (CD44) and display high aldehyde dehydrogenase activity that converts vitamin A-derived retinal into retinoic acids. All-trans retinoic acid (ATRA), which has pro-differentiating properties, has revolutionized the prognosis of acute promyelotic leukemia by increasing its remission rate from 15% to 85%. Recent studies have started to show that ATRA also has an anti-tumoral role on solid cancers such as GC. The purpose of this review is therefore to summarize the work that evaluated the effects of ATRA in GC and to evaluate whether its anti-cancerous action involves gastric CSCs targeting. It has been demonstrated that ATRA can block the cell cycle, enhance apoptosis, and decrease gastric CSCs properties in GC cell lines, tumorspheres, and patient-derived xenograft mice models. Therefore, retinoids and new synthetic retinoids seem to be a promising step forward in targeted therapy of gastric CSC in combination with existing chemotherapies. Future studies should probably focus on these points.
Collapse
Affiliation(s)
- Damien Bouriez
- INSERM, U1053, Bordeaux Research in Translational Oncology, 33000 Bordeaux, France.
- Department of Digestive Surgery, Haut-Lévêque Hospital, 33000 Bordeaux, France.
| | - Julie Giraud
- INSERM, U1053, Bordeaux Research in Translational Oncology, 33000 Bordeaux, France.
- Department of Life and Health Sciences, University of Bordeaux, 33000 Bordeaux, France.
| | - Caroline Gronnier
- INSERM, U1053, Bordeaux Research in Translational Oncology, 33000 Bordeaux, France.
- Department of Digestive Surgery, Haut-Lévêque Hospital, 33000 Bordeaux, France.
- Department of Life and Health Sciences, University of Bordeaux, 33000 Bordeaux, France.
| | - Christine Varon
- INSERM, U1053, Bordeaux Research in Translational Oncology, 33000 Bordeaux, France.
- Department of Life and Health Sciences, University of Bordeaux, 33000 Bordeaux, France.
| |
Collapse
|
19
|
Xiang Z, Huang X, Wang J, Zhang J, Ji J, Yan R, Zhu Z, Cai W, Yu Y. Cross-Database Analysis Reveals Sensitive Biomarkers for Combined Therapy for ERBB2+ Gastric Cancer. Front Pharmacol 2018; 9:861. [PMID: 30123134 PMCID: PMC6085474 DOI: 10.3389/fphar.2018.00861] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/17/2018] [Indexed: 12/13/2022] Open
Abstract
Exploring ERBB2-related pathways will help us finding sensitive molecules and potential combined therapeutic targets of ERBB2-targeted therapy for ERBB2+ gastric cancer (GC). In this study, we performed a cross-databases study focused on ERBB2+ GC. The data of ERBB2+ GC deposited in the cancer genome atlas (TCGA), gene expression omnibus (GEO), InBio MapTM, cancer cell line encyclopedia (CCLE), and cancer therapeutics response portal (CTRP) were analyzed. The correlation of expression levels of candidate and IC50 of candidate genes-targeted drugs were verified on NCI-N87 and MKN-45 GC cell lines. We found that RARA, THRA, CACNB1, and TOP2A are drug sensitive biomarkers of ERBB2-targeted treatment with FDA-approved drugs. All these genes act through Myc signaling pathway. Myc is the downstream hub gene of both ERBB2 and RARA. The expression of RARA, THRA, and CACNB1 were negatively correlated with Myc activation, while ERBB2 and TOP2A positively correlated with Myc activation. SH3BGRL3, SH3BGRL, and NRG2 were identified as potential ligands of ERBB2. The ERBB2+ GC with RARA amplification demonstrated better prognosis than those without RARA amplification, while overexpression of NRG2 and SH3BGRL correlated with poor prognosis in ERBB2+ GC. About 90% of ERBB2+ GC was compatible with chromosome instability (CIN) subtype of TCGA, which overlaps with intestinal-type GC in Lauren classification. In validating experiments, combination of Lapatinib and all-trans retinoic acid (ATRA) synergistically suppresses cell growth, and accompanied by decreased expression of MYC. In conclusions, we identified several predicting biomarkers for ERBB2-targeted therapy and corresponding histological features of ERBB2+ GC. Combination of ERBB2 antagonist or RARA agonist may be effective synergistic regimens for ERBB2+ GC.
Collapse
Affiliation(s)
- Zhen Xiang
- Department of Surgery, Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai, China
| | - Xia Huang
- Department of Disease Prevention and Control, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiexuan Wang
- Department of Surgery, Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai, China
| | - Jun Zhang
- Department of Surgery, Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai, China
| | - Jun Ji
- Department of Surgery, Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai, China
| | - Ranlin Yan
- Department of Surgery, Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai, China
| | - Zhenggang Zhu
- Department of Surgery, Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai, China
| | - Wei Cai
- Department of Surgery, Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai, China
| | - Yingyan Yu
- Department of Surgery, Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai, China
| |
Collapse
|
20
|
Specific inhibitor of Notch‑3 enhances the sensitivity of NSCLC cells to gemcitabine. Oncol Rep 2018; 40:155-164. [PMID: 29781034 PMCID: PMC6059738 DOI: 10.3892/or.2018.6448] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 05/03/2018] [Indexed: 12/18/2022] Open
Abstract
Notch-3 is a receptor of the Notch signaling pathway and plays an important role in regulating self-renewal, differentiation and apoptosis in cancer cells. Overexpression of Notch-3 has been proved to be associated with resistance to gemcitabine (GEM) and poor patient prognosis for various malignant tumors. In the present study, two non-small cell lung cancer (NSCLC) cell lines, H1299 and A549, were induced with GEM for two months and then were treated with various concentrations of a Notch signaling blocker, N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT), with the goal of reducing expression of Notch intracellular domain 3 (NICD3). Both cell lines were subsequently treated with either DAPT or DAPT combined with GEM and then viability, apoptosis, colony formation and cell count assays were performed. DAPT treatment effectively downregulated the expression of NICD3 in both cell lines. DAPT combined with GEM also significantly reduced the percentage of viable cells in both cell lines, while increasing the percentage of apoptotic cells, compared with GEM alone. In the clonogenicity assays, the combination of DAPT and GEM led to a decrease in clone numbers and significantly greater inhibition of the H1299 and A549 cells compared to treatment with DAPT or GEM alone. Meanwhile, levels of the apoptosis-related proteins, Bcl-2 and Bax, were found to be affected by the various treatments. Thus Notch-3 appears to be a promising target for gene therapy and DAPT is able to mediate a strong antitumor effect in NSCLC cells that overexpress Notch-3. Further studies of a combined treatment regimen with DAPT and GEM are warranted and may provide greater efficacy and safety in the treatment of NSCLC patients.
Collapse
|