1
|
Badreldin H, Elshal M, El-Karef A, Ibrahim T. Empagliflozin protects the heart from atrial fibrillation in rats through inhibiting the NF-κB/HIF-1α regulatory axis and atrial remodeling. Int Immunopharmacol 2024; 143:113403. [PMID: 39437485 DOI: 10.1016/j.intimp.2024.113403] [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: 04/18/2024] [Revised: 07/04/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
Atrial fibrillation (AF) is the most common form of sustained cardiac arrhythmia. The current study aimed to investigate the potential of empagliflozin (EMPA) to protect against acetylcholine (ACh)/calcium chloride (CaCl2)-induced AF in rats and elucidate the possible underlying mechanism of action. Rats were randomly assigned to five groups, as follows: CTRL group: received 1 ml/kg isotonic saline; AF group: received 1 ml/kg induction mixture of ACh/CaCl2 (60 µg ACh and 10 mg CaCl2 per ml); EMPA group: received 30 mg/kg EMPA; AF + EMPA10 group: received the induction mixture concurrent with 10 mg/kg EMPA; AF + EMPA30 group: received the induction mixture concurrent with 30 mg/kg EMPA. Our results showed that EMPA administration inhibited the AF-related electrocardiographic abnormalities and decreased the serum brain natriuretic peptide levels. EMPA treatment maintained the cardiac redox balance, as indicated by reduced levels of the lipid peroxidation biomarker malonaldehyde while enhancing the antioxidant glutathione levels. Moreover, EMPA markedly repressed ACh/CaCl2-induced C-reactive protein, tumor necrosis factor, and interleukin-6 production. Interestingly, EMPA administration strongly suppressed cardiac transforming growth factor beta1, collagen type I, and alpha-smooth muscle actin expression levels in the AF rats. These results were consistent with our histopathological findings, which revealed the ameliorative effect of EMPA on AF-induced inflammatory and fibrotic lesions. Mechanistically, EMPA dose-dependently downregulated nuclear factor-kappa B (NF-κB) and hypoxia-inducible factor (HIF)-1α expressions. Besides, it attenuated the pro-apoptotic active caspase-3 while augmenting the anti-apoptotic B-cell lymphoma 2 expressions. Furthermore, EMPA dose-dependently suppressed cardiac phosphatidylinositol 3-kinase (PI3K)/Akt signaling. In conclusion, this study demonstrates that EMPA intervention, within AF induction, protects against ACh/CaCl2-induced AF in rats, exerting powerful antioxidant, anti-inflammatory, anti-fibrotic, and anti-apoptotic effects. These effects are mainly mediated through the targeting of the NF-κB/HIF-1α regulatory axis in a dose-dependent manner.
Collapse
Affiliation(s)
- Hussein Badreldin
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Egypt
| | - Mahmoud Elshal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Egypt.
| | - Amr El-Karef
- Department of Pathology, Faculty of Medicine, Mansoura University, Egypt; Department of Pathology, Faculty of Medicine, Horus University, Egypt
| | - Tarek Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Egypt
| |
Collapse
|
2
|
Wilczyński JR, Wilczyński M, Paradowska E. Cancer Stem Cells in Ovarian Cancer-A Source of Tumor Success and a Challenging Target for Novel Therapies. Int J Mol Sci 2022; 23:ijms23052496. [PMID: 35269636 PMCID: PMC8910575 DOI: 10.3390/ijms23052496] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 02/04/2023] Open
Abstract
Ovarian cancer is the most lethal neoplasm of the female genital organs. Despite indisputable progress in the treatment of ovarian cancer, the problems of chemo-resistance and recurrent disease are the main obstacles for successful therapy. One of the main reasons for this is the presence of a specific cell population of cancer stem cells. The aim of this review is to show the most contemporary knowledge concerning the biology of ovarian cancer stem cells (OCSCs) and their impact on chemo-resistance and prognosis in ovarian cancer patients, as well as to present the treatment options targeted exclusively on the OCSCs. The review presents data concerning the role of cancer stem cells in general and then concentrates on OCSCs. The surface and intracellular OCSCs markers and their meaning both for cancer biology and clinical prognosis, signaling pathways specifically activated in OCSCs, the genetic and epigenetic regulation of OCSCs function including the recent studies on the non-coding RNA regulation, cooperation between OCSCs and the tumor microenvironment (ovarian cancer niche) including very specific environment such as ascites fluid, the role of shear stress, autophagy and metabolic changes for the function of OCSCs, and finally mechanisms of OCSCs escape from immune surveillance, are described and discussed extensively. The possibilities of anti-OCSCs therapy both in experimental settings and in clinical trials are presented, including the recent II phase clinical trials and immunotherapy. OCSCs are a unique population of cancer cells showing a great plasticity, self-renewal potential and resistance against anti-cancer treatment. They are responsible for the progression and recurrence of the tumor. Several completed and ongoing clinical trials have tested different anti-OCSCs drugs which, however, have shown unsatisfactory efficacy in most cases. We propose a novel approach to ovarian cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Jacek R Wilczyński
- Department of Gynecological Surgery and Gynecological Oncology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
- Correspondence:
| | - Miłosz Wilczyński
- Department of Gynecological, Endoscopic and Oncological Surgery, Polish Mother’s Health Center—Research Institute, 281/289 Rzgowska Str., 93-338 Lodz, Poland;
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, 4 Kosciuszki Str., 90-419 Lodz, Poland
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, 106 Lodowa Str., 93-232 Lodz, Poland;
| |
Collapse
|
3
|
Jiang Q, Geng X, Warren J, Eugene Paul Cosky E, Kaura S, Stone C, Li F, Ding Y. Hypoxia Inducible Factor-1α (HIF-1α) Mediates NLRP3 Inflammasome-Dependent-Pyroptotic and Apoptotic Cell Death Following Ischemic Stroke. Neuroscience 2020; 448:126-139. [PMID: 32976985 DOI: 10.1016/j.neuroscience.2020.09.036] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 12/19/2022]
Abstract
Stroke is a major cause of death and long-term disability. Recent evidence suggests that hypoxia-inducible factor 1α (HIF-1α), a transcription factor that regulates oxygen levels, plays a key role in neurological outcomes after ischemic stroke. Accordingly, we investigated the mechanism of HIF-1α on pyroptotic and apoptotic cells during ischemia/reperfusion (I/R). Adult Sprague-Dawley rats underwent 2 h of middle cerebral artery occlusion (MCAO). The rats were then exposed to 6 or 24 h of reperfusion, with or without YC-1 (HIF-1α inhibitor, 5 mg/kg). Infarct volumes, along with mRNA and protein quantities of HIF-1α, NLRP3, IL-1β, IL-18, Caspase-1, and co-localization of HIF-1α, and NLRP3, were assessed. We measured apoptotic and pyroptotic cell death, gasdermin D (GSDMD) activation and lactate dehydrogenase (LDH) activity, and the infiltration of neutrophils and macrophages after ischemic stroke. HIF-1α mRNA and NLRP3 inflammasome components were increased after 24 h of reperfusion. YC-1 significantly reduced the mRNA and protein expression of NLRP3, IL-1β, IL-18, and caspase-1; significantly decreased infarction and pyroptotic cell death after 24 h of reperfusion; attenuated the neuroinflammatory response by reducing infiltration of CD68- and MPO-positive cells after 24 h of reperfusion; and reduced apoptotic cell death following ischemic stroke. We found that HIF-1α likely regulates inflammatory responses through the NLRP3 inflammasome complex, thus influencing both apoptotic and pyroptotic cell death after stroke. These findings suggest that future investigations are needed regarding HIF-1α and its role as a potential molecular target in the treatment of acute ischemic stroke.
Collapse
Affiliation(s)
- Qian Jiang
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China; Department of Neurology, Beijing Luhe Hospital, Capital Medical University, China
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China; Department of Neurology, Beijing Luhe Hospital, Capital Medical University, China; Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Jonathan Warren
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | | | - Shawn Kaura
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Christopher Stone
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Fengwu Li
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, China; Department of Neurology, Beijing Luhe Hospital, Capital Medical University, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA; Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI, USA.
| |
Collapse
|
4
|
Bhukya PL, Laxmivandana R, Sundaram GM. NF-κB Role and Potential Drug Targets in Gastrointestinal Cancer. ROLE OF TRANSCRIPTION FACTORS IN GASTROINTESTINAL MALIGNANCIES 2017:45-71. [DOI: 10.1007/978-981-10-6728-0_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
5
|
Li S, Wei Q, Li Q, Zhang B, Xiao Q. Down-regulating HIF-1α by lentivirus-mediated shRNA for therapy of triple negative breast cancer. Cancer Biol Ther 2016; 16:866-75. [PMID: 25920936 DOI: 10.1080/15384047.2015.1040958] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Hypoxia is associated with poor response to treatment in various cancers. Hypoxia inducible factor 1 (HIF-1) is a major transcription factor that mediates adaptation of cancer cells to a hypoxic environment and regulates many genes that are involved in key cellular functions, including cell immortalization, stem cell maintenance, autocrine growth/survival, angiogenesis, invasion/metastasis, and resistance to chemotherapy. HIF-1α has been considered as an attractive therapeutic target for cancer treatment, but there is limited success in this research field. In the present study, we designed a recombinant lentivirus containing HIF-1α siRNA, developed stably transfected cell lines, and tested the anticancer effects of the siRNA on cancer cells in vitro and in vivo. Our results indicated that the stable downregulation of HIF-1α reversed chemoresistance, inhibited proliferation, migration and invasion of cancer cells, and slowed down the tumor growth in breast cancer xenograft models. In conclusion, the recombinant lentivirus containing HIF-1α siRNA provides a new avenue for developing novel therapy for triple negative breast cancer.
Collapse
Affiliation(s)
- Shuang Li
- a Graduate School of Southern Medical University ; Guangzhou , China
| | | | | | | | | |
Collapse
|
6
|
Wozniak M, Sztiller-Sikorska M, Czyz M. Diminution of miR-340-5p levels is responsible for increased expression of ABCB5 in melanoma cells under oxygen-deprived conditions. Exp Mol Pathol 2015; 99:707-16. [PMID: 26554847 DOI: 10.1016/j.yexmp.2015.11.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 11/06/2015] [Accepted: 11/06/2015] [Indexed: 12/21/2022]
Abstract
Melanoma is usually highly refractory to chemotherapy. This resistance to treatment is mainly due to high heterogeneity and plasticity of melanoma cells strictly connected to changes in tumor microenvironment. Hypoxia can drastically alter cancer biology. Solid tumor cells under hypoxia gain stem-like features, they are more invasive and drug-resistant than their normoxic counterparts. These effects could be mediated by changes in miRNA expression under hypoxia. MiRNAs are small non-coding RNA molecules that can negatively control gene expression. In the present study using microarray technology we evaluated the expression of miRNAs in melanoma cells derived from nodular melanoma and grown under normoxic and hypoxic conditions. Using R environment for statistical analysis we found that 70 miRNAs were differentially-expressed, and 16 of them were significantly down-regulated in melanoma cells grown in hypoxic conditions compared to cells grown in normoxia. We intended to find transcripts whose expression is increased due to down-regulation of selected miRNAs. Bioinformatics analysis revealed that increased levels of HIF-2α, ABCB5, OCT4, SOX2 and ZEB1 in different melanoma populations under hypoxia could be a result of significant down-regulation of miR-340-5p. Inhibition of miR-340-5p confirmed that this miRNA negatively influences the expression of ABCB5. This is the first study showing the relationship between miR-340-5p and expression of ABCB5, a transmembrane transporter involved in drug resistance considered as a marker of melanoma stem-like cells.
Collapse
Affiliation(s)
- Michal Wozniak
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215 Lodz, Poland.
| | - Malgorzata Sztiller-Sikorska
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215 Lodz, Poland
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215 Lodz, Poland
| |
Collapse
|
7
|
Cui H, Seubert B, Stahl E, Dietz H, Reuning U, Moreno-Leon L, Ilie M, Hofman P, Nagase H, Mari B, Krüger A. Tissue inhibitor of metalloproteinases-1 induces a pro-tumourigenic increase of miR-210 in lung adenocarcinoma cells and their exosomes. Oncogene 2014; 34:3640-50. [DOI: 10.1038/onc.2014.300] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 07/30/2014] [Accepted: 07/31/2014] [Indexed: 12/12/2022]
|
8
|
Tang FR, Loke WK. Molecular mechanisms of low dose ionizing radiation-induced hormesis, adaptive responses, radioresistance, bystander effects, and genomic instability. Int J Radiat Biol 2014; 91:13-27. [DOI: 10.3109/09553002.2014.937510] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
9
|
Chen F, Liu Y, Wang S, Guo X, Shi P, Wang W, Xu B. Triptolide, a Chinese herbal extract, enhances drug sensitivity of resistant myeloid leukemia cell lines through downregulation of HIF-1α and Nrf2. Pharmacogenomics 2014; 14:1305-17. [PMID: 23930677 DOI: 10.2217/pgs.13.122] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
AIM To explore whether triptolide (TPL) can enhance drug sensitivity of resistant myeloid leukemia cell lines through downregulation of HIF-1α and Nrf2. MATERIALS & METHODS HL60/A and K562/G cells were subjected to different treatments and thereafter an methyl thiazole tetrazolium bromide assay, flow cytometry, western blot and real-time PCR were used to determine IC₅₀, apoptotic status and expression of Nrf2, HIF-1α and their target genes. RESULTS Doxorubicin- or imatinib-induced apoptosis was enhanced when anticancer agents were used in combination with TPL. When combined with TPL, both doxorubicin and imatinib downregulate Nrf2 and HIF-1α expression at protein and mRNA levels. Genes downstream of Nrf2, for example, NQO1, GSR and HO-1, as well as target genes of HIF-1α, for example, BNIP3, VEGF and CAIX are also downregulated at the mRNA level. CONCLUSION TPL is able to enhance drug sensitivity of resistant myeloid leukemia cell lines through downregulation of HIF-1α and Nrf2.
Collapse
Affiliation(s)
- Feili Chen
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | | | | | | | | | | | | |
Collapse
|
10
|
Gauter-Fleckenstein B, Reboucas JS, Fleckenstein K, Tovmasyan A, Owzar K, Jiang C, Batinic-Haberle I, Vujaskovic Z. Robust rat pulmonary radioprotection by a lipophilic Mn N-alkylpyridylporphyrin, MnTnHex-2-PyP(5+). Redox Biol 2014; 2:400-10. [PMID: 24624330 PMCID: PMC3949096 DOI: 10.1016/j.redox.2013.12.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 12/19/2013] [Accepted: 12/20/2013] [Indexed: 12/21/2022] Open
Abstract
With the goal to enhance the distribution of cationic Mn porphyrins within mitochondria, the lipophilic Mn(III)meso-tetrakis(N-n-hexylpyridinium-2-yl)porphyrin, MnTnHex-2-PyP(5+) has been synthesized and tested in several different model of diseases, where it shows remarkable efficacy at as low as 50 µg/kg single or multiple doses. Yet, in a rat lung radioprotection study, at higher 0.6-1 mg/kg doses, due to its high accumulation and micellar character, it became toxic. To avoid the toxicity, herein the pulmonary radioprotection of MnTnHex-2-PyP(5+) was assessed at 50 µg/kg. Fischer rats were irradiated to their right hemithorax (28 Gy) and treated with 0.05 mg/kg/day of MnTnHex-2-PyP(5+) for 2 weeks by subcutaneously-implanted osmotic pumps, starting at 2 h post-radiation. The body weights and breathing frequencies were followed for 10 weeks post-radiation, when the histopathology and immunohistochemistry were assessed. Impact of MnTnHex-2-PyP(5+) on macrophage recruitment (ED-1), DNA oxidative damage (8-OHdG), TGF-β1, VEGF(A) and HIF-1α were measured. MnTnHex-2-PyP(5+) significantly decreased radiation-induced lung histopathological (H&E staining) and functional damage (breathing frequencies), suppressed oxidative stress directly (8-OHdG), or indirectly, affecting TGF-β1, VEGF (A) and HIF-1α pathways. The magnitude of the therapeutic effects is similar to the effects demonstrated under same experimental conditions with 120-fold higher dose of ~5000-fold less lipophilic Mn(III)meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin, MnTE-2-PyP(5+).
Collapse
Key Words
- 8-OHdG, 8-hydroxy-2'-deoxyguanosine
- AKT, protein kinase B (PKB), a serine/threonine-specific protein kinase
- ALS, amyotrophic laterial sclerosis
- AP-1, activator protein-1
- AT, ataxia telangiectasia
- BBB, blood brain barrier
- Breathing frequencies
- CNS, central nervous system
- CO3−, carbonate radical
- ClO−, hypochlorite
- ETC, mitochondrial electron transport chain
- Fischer rats
- GMP, good manufacturing practice
- GS−, monodeprotonated glutathione
- HIF-1α, hypoxia inducible factor-1
- HO2−, monodeprotonated hydrogen peroxide
- Histopathology
- I/R, ischemia reperfusion
- Immunohistochemistry
- Lung injury
- MCAO, middle cerebral artery occlusion
- Manganese porphyrins
- MnP, Mn porphyrin
- MnTDE-2-ImP5+, Mn(III) tetrakis[N,N'-diethylimidazolium-2-yl)porphyrin, AEOL10150
- MnTE-2-PyP5+
- MnTE-2-PyP5+, Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin (AEOL10113)
- MnTnBuOE-2-PyP5+, Mn(III) meso-tetrakis(N-(n-butoxyethyl)pyridinium-2-yl)porphyrin
- MnTnHex-2-PyP5+
- MnTnHex-2-PyP5+, Mn(III) meso-tetrakis(N-(n-hexyl)pyridinium-2-yl)porphyrin (AEOL10113)
- NF-κB, nuclear factor κB
- NHE, normal hydrogen electrode
- NO, nitric oxide
- NOX4, NADPH oxidase, isoform 4 E1/2, Half-wave metal-centered reduction potential
- Nrf-2, nuclear factor-erythroid-derived 2-like 2
- O2−, superoxide
- ONOO−, peroxynitrite
- PI3K, phosphatidylinositide 3-kinase
- PTEN, phosphoinositide 3-phosphatase
- Radioprotection
- Redox-modulators
- SAH, subarachnoid hemorrhage
- SOD, superoxide dismutase
- SP-1, specificity protein-1
- TF, transcription factor
- TGF-β1, one of the 3 members of the TGF-β transforming growth factor-β family
- VEGF, vascular endothelial growth factor
- mTOR, mammalian target of rapamycin (mTOR), a serine/threonine protein kinase
Collapse
Affiliation(s)
- Benjamin Gauter-Fleckenstein
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA ; Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Julio S Reboucas
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Katharina Fleckenstein
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA ; Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kouros Owzar
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, USA ; Biostatistics and Computational Biology Core, RadCCORE, Duke University Medical Center, Durham, USA
| | - Chen Jiang
- Biostatistics and Computational Biology Core, RadCCORE, Duke University Medical Center, Durham, USA
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA ; Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland, 655W Baltimore Street, Bressler Research Building, 8-025, Baltimore, MD 21201, USA
| |
Collapse
|
11
|
Low-dose triptolide in combination with idarubicin induces apoptosis in AML leukemic stem-like KG1a cell line by modulation of the intrinsic and extrinsic factors. Cell Death Dis 2013; 4:e948. [PMID: 24309935 PMCID: PMC3877540 DOI: 10.1038/cddis.2013.467] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 10/17/2013] [Accepted: 10/21/2013] [Indexed: 01/20/2023]
Abstract
Leukemia stem cells (LSCs) are considered to be the main reason for relapse and are also regarded as a major hurdle for the success of acute myeloid leukemia chemotherapy. Thus, new drugs targeting LSCs are urgently needed. Triptolide (TPL) is cytotoxic to LSCs. Low dose of TPL enhances the cytotoxicity of idarubicin (IDA) in LSCs. In this study, the ability of TPL to induce apoptosis in leukemic stem cell (LSC)-like cells derived from acute myeloid leukemia cell line KG1a was investigated. LSC-like cells sorted from KG1a were subjected to cell cycle analysis and different treatments, and then followed by in vitro methyl thiazole tetrazolium bromide cytotoxicity assay. The effects of different drug combinations on cell viability, intracellular reactive-oxygen species (ROS) activity, colony-forming ability and apoptotic status were also examined. Combination index-isobologram analysis indicates a synergistic effect between TPL and IDA, which inhibits the colony-forming ability of LSC-like cells and induces their apoptosis. We further investigated the expression of Nrf2, HIF-1α and their downstream target genes. LSC-like cells treated with both TPL and IDA have increased levels of ROS, decreased expression of Nrf2 and HIF-1α pathways. Our findings indicate that the synergistic cytotoxicity of TPL and IDA in LSCs-like cells may attribute to both induction of ROS and inhibition of the Nrf2 and HIF-1α pathways.
Collapse
|
12
|
In papillary thyroid carcinoma, TIMP-1 expression correlates with BRAF V600E mutation status and together with hypoxia-related proteins predicts aggressive behavior. Virchows Arch 2013; 463:437-44. [DOI: 10.1007/s00428-013-1453-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 06/26/2013] [Accepted: 07/08/2013] [Indexed: 12/29/2022]
|
13
|
McNamee EN, Korns Johnson D, Homann D, Clambey ET. Hypoxia and hypoxia-inducible factors as regulators of T cell development, differentiation, and function. Immunol Res 2013; 55:58-70. [PMID: 22961658 DOI: 10.1007/s12026-012-8349-8] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Oxygen is a molecule that is central to cellular respiration and viability, yet there are multiple physiologic and pathological contexts in which cells experience conditions of insufficient oxygen availability, a state known as hypoxia. Given the metabolic challenges of a low oxygen environment, hypoxia elicits a range of adaptive responses at the cellular, tissue, and systemic level to promote continued survival and function. Within this context, T lymphocytes are a highly migratory cell type of the adaptive immune system that frequently encounters a wide range of oxygen tensions in both health and disease. It is now clear that oxygen availability regulates T cell differentiation and function, a response orchestrated in large part by the hypoxia-inducible factor transcription factors. Here, we discuss the physiologic scope of hypoxia and hypoxic signaling, the contribution of these pathways in regulating T cell biology, and current gaps in our understanding. Finally, we discuss how emerging therapies that modulate the hypoxic response may offer new modalities to alter T cell function and the outcome of acute and chronic pathologies.
Collapse
Affiliation(s)
- Eóin N McNamee
- Mucosal Inflammation Program, Department of Anesthesiology, School of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | | | | | | |
Collapse
|
14
|
Sun J, Zhang D, Bae DH, Sahni S, Jansson P, Zheng Y, Zhao Q, Yue F, Zheng M, Kovacevic Z, Richardson DR. Metastasis suppressor, NDRG1, mediates its activity through signaling pathways and molecular motors. Carcinogenesis 2013; 34:1943-54. [PMID: 23671130 DOI: 10.1093/carcin/bgt163] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The metastasis suppressor, N-myc downstream regulated gene 1 (NDRG1), is negatively correlated with tumor progression in multiple neoplasms, being a promising new target for cancer treatment. However, the precise molecular effects of NDRG1 remain unclear. Herein, we summarize recent advances in understanding the impact of NDRG1 on cancer metastasis with emphasis on its interactions with the key oncogenic nuclear factor-kappaB, phosphatidylinositol-3 kinase/phosphorylated AKT/mammalian target of rapamycin and Ras/Raf/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase signaling pathways. Recent studies demonstrating the inhibitory effects of NDRG1 on the epithelial-mesenchymal transition, a key initial step in metastasis, TGF-β pathway and the Wnt/β-catenin pathway are also described. Furthermore, NDRG1 was also demonstrated to regulate molecular motors in cancer cells, leading to inhibition of F-actin polymerization, stress fiber formation and subsequent reduction of cancer cell migration. Collectively, this review summarizes the underlying molecular mechanisms of the antimetastatic effects of NDRG1 in cancer cells.
Collapse
Affiliation(s)
- Jing Sun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Xiao LJ, Chen YY, Lin P, Zou HF, Lin F, Zhao LN, Li D, Guo L, Tang JB, Zheng XL, Yu XG. Hypoxia increases CX3CR1 expression via HIF-1 and NF‑κB in androgen-independent prostate cancer cells. Int J Oncol 2012; 41:1827-36. [PMID: 22941344 DOI: 10.3892/ijo.2012.1610] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 05/25/2012] [Indexed: 11/06/2022] Open
Abstract
The unique CX3C chemokine CX3CL1 and its cognate receptor CX3CR1 have been implicated in organ-specific metastasis of various types of tumors. Hypoxia, a common phenomenon in solid tumors, is associated with a malignant cancer phenotype. Previous studies have proved that hypoxia facilitates cancer cell metastasis through upregulation of specific chemokine receptors. We hypothesized that hypoxia could upregulate CX3CR1 expression and lead to an increased chemotactic response to CX3CL1 in prostate cancer cells. In the present study, we found that CX3CR1 expression was significantly increased in androgen-independent prostate cancer cells, including DU145, PC-3 and PC-3M, following exposure to hypoxia. This upregulation of CX3CR1 corresponded to a significant increase in migration and invasion of prostate cancer cells under hypoxic conditions, which was attenuated after knocking down CX3CR1 expression. In addition, we examined the possible role of HIF-1 and NF-κB in the process of hypoxia-induced CX3CR1 expression and hypoxia-mediated metastasis. Attenuation of HIF-1 and NF-κB transcriptional activity by siRNAs or pharmacological inhibitors, abrogated hypoxia-induced upregulation of CX3CR1, and also prevented the migration and invasion of DU145 cells under a hypoxic environment. In summary, our study demonstrated that HIF-1 and NF-κB are essential for hypoxia-regulated CX3CR1 expression, which is associated with increased migratory and invasive potential of prostate cancer cells. CX3CR1 signaling is a potential therapeutic target in the adjuvant treatment of prostate cancer.
Collapse
Affiliation(s)
- Li-Jie Xiao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Liu HT, Wang LL, Liu LX. Advances in understanding mechanisms underlying the antitumor activity of curcumin analogue EF24. Shijie Huaren Xiaohua Zazhi 2012; 20:1853-1857. [DOI: 10.11569/wcjd.v20.i20.1853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Curcumin, a natural polyphenol which was first extracted by Vogel and Pelletier from rhizomes of the plant Curcuma longa L, has potent anticarcinogenic activity and low toxic side effects in a wide variety of tumor cells. It has been listed as a third-generation chemoprophylactic drug by the US National Cancer Institute. However, the therapeutic benefit is hampered by its low absorption after transdermal or oral application. Ames et al. have developed a series of novel synthetic curcumin analogs that are more potent and have better water solubility than curcumin. One of these leading compounds, EF24, exhibits about 10-fold greater cytotoxic activity against various cancer cell lines in relation to curcumin. This article will review the recent advances in understanding mechanisms underlying the antitumor activity of EF24.
Collapse
|
17
|
Cui H, Grosso S, Schelter F, Mari B, Krüger A. On the Pro-Metastatic Stress Response to Cancer Therapies: Evidence for a Positive Co-Operation between TIMP-1, HIF-1α, and miR-210. Front Pharmacol 2012; 3:134. [PMID: 22807917 PMCID: PMC3395024 DOI: 10.3389/fphar.2012.00134] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 06/24/2012] [Indexed: 01/18/2023] Open
Abstract
In contrast to expectations in the past that tumor starvation or unselective inhibition of proteolytic activity would cure cancer, there is accumulating evidence that microenvironmental stress, such as hypoxia or broad-spectrum inhibition of metalloproteinases can promote metastasis. In fact, malignant tumor cells, due to their genetic and epigenetic instability, are predisposed to react to stress by adaptation and, if the stress persists, by escape and formation of metastasis. Recent recognition of the concepts of dynamic evolution as well as population and systems biology is extremely helpful to understand the disappointments of clinical trials with new drugs and may lead to paradigm-shifts in therapy strategies. This must be complemented by an increased understanding of molecular mechanism involved in stress response. Here, we review new roles of Hypoxia-inducible factor-1 (HIF-1), one transcription factor regulating stress response-related gene expression: HIF-1 is crucial for invasion and metastasis, independent from its pro-survival function. In addition, HIF-1 mediates pro-metastatic microenvironmental changes of the proteolytic balance as triggered by high systemic levels of tissue inhibitor of metalloproteinases-1 (TIMP-1), typical for many aggressive cancers, and regulates the metabolic switch to glycolysis, notably via activation of the microRNA miR-210. There is preliminary evidence that TIMP-1 also induces miR-210. Such positive-regulatory co-operation of HIF-1α, miR-210, and TIMP-1, all described to correlate with bad prognosis of cancer patients, opens new perspectives of gaining insight into molecular mechanisms of metastasis-inducing evasion of tumor cells from stress.
Collapse
Affiliation(s)
- Haissi Cui
- Klinikum Rechts der Isar der Technischen Universität München, Institut für Experimentelle Onkologie und Therapieforschung München, Germany
| | | | | | | | | |
Collapse
|