1
|
Mansour AM, Khaled RM, Ferraro G, Shehab OR, Merlino A. Metal-based carbon monoxide releasing molecules with promising cytotoxic properties. Dalton Trans 2024; 53:9612-9656. [PMID: 38808485 DOI: 10.1039/d4dt00087k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Carbon monoxide, the "silent killer" gas, is increasingly recognised as an important signalling molecule in human physiology, which has beneficial biological properties. A particular way of achieving controlled CO administration is based on the use of biocompatible molecules that only release CO when triggered by internal or external factors. These approaches include the development of pharmacologically effective prodrugs known as CO releasing molecules (CORMs), which can supply biological systems with CO in well-regulated doses. An overview of transition metal-based CORMs with cytotoxic properties is here reported. The mechanisms at the basis of the biological activities of these molecules and their potential therapeutical applications with respect to their stability and CO releasing properties have been discussed. The activation of metal-based CORMs is determined by the type of metal and by the nature and features of the auxiliary ligands, which affect the metal core electronic density and therefore the prodrug resistance towards oxidation and CO release ability. A major role in regulating the cytotoxic properties of these CORMs is played by CO and/or CO-depleted species. However, several mysteries concerning the cytotoxicity of CORMs remain as intriguing questions for scientists.
Collapse
Affiliation(s)
- Ahmed M Mansour
- Department of Chemistry, United Arab Emirates University, Al-Ain, United Arab Emirates.
| | - Rabaa M Khaled
- Department of Chemistry, Faculty of Science, Cairo University, Gamma Street, 12613, Egypt.
| | - Giarita Ferraro
- Department of Chemical Sciences, University of Naples Federico II, Napoli, Italy.
| | - Ola R Shehab
- Department of Chemistry, Faculty of Science, Cairo University, Gamma Street, 12613, Egypt.
| | - Antonello Merlino
- Department of Chemical Sciences, University of Naples Federico II, Napoli, Italy.
| |
Collapse
|
2
|
Rueda Huélamo MA, Martínez Perlado A, Consoli V, García-Tejedor A, Haros CM, Laparra Llopis JM. Improvement of hepatic innate immunity in chemically-injured livers to develop hepatocarcinoma by a serine type-protease inhibitors enriched extract from Chenopodium quinoa. Food Funct 2024; 15:3600-3614. [PMID: 38469889 DOI: 10.1039/d3fo03083k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Food ingredients have critical effects on the maturation and development of the immune system, which innate - lymphoid (ILCs) and myeloid - cells play key roles as important regulators of energy storage and hepatic fat accumulation. Therefore, the objective of this study is to define potential links between a dietary immunonutritional induction of the selective functional differentiation of monocytes-derived macrophages, ILCs and lipid homeostasis in hepatocarcinoma (HCC)-developing mice. Hepatic chemically injured (diethylnitrosamine/thiacetamide) Rag2-/- and Rag2-/-Il2-/- mice were administered with serine-type protease inhibitors (SETIs) obtained from Chenopodium quinoa. Early HCC-driven immunometabolic imbalances (infiltrated macrophages, glucose homeostasis, hepatic lipid profile, ILCs expansion, inflammatory conditions, microbiota) in animals put under a high-fat diet for 2 weeks were assessed. It was also approached the potential of SETIs to cause functional adaptations of the bioenergetics of human macrophage-like cells (hMLCs) in vitro conditioning their capacity to accumulate fat. It is showed that Rag2-/-Il2-/- mice, lacking ILCs, are resistant to the SETIs-induced hepatic macrophages (CD68+F4/80+) activation. Feeding SETIs to Rag2-/- mice, carrying ILCs, promoted the expansion towards ILC3s (CD117+Nkp46+CD56+) and reduced that of ILC2s (CD117+KLRG1+) into livers. In vitro studies demonstrate that hMLCs, challenged to SETIs, develop a similar phenotype of that found in mice and bioenergetic adaptations leading to increased lipolysis. It is concluded that SETIs promote liver macrophage activation and ILCs adaptations to ameliorate HCC-driven immunometabolic imbalances.
Collapse
Affiliation(s)
- Maria Alicia Rueda Huélamo
- Molecular Immunonutrition Group, Madrid Institute for Advanced Studies in Food (IMDEA-Food), Madrid, Spain.
| | - Alba Martínez Perlado
- Molecular Immunonutrition Group, Madrid Institute for Advanced Studies in Food (IMDEA-Food), Madrid, Spain.
| | - Valeria Consoli
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria, 6, Catania 95125, Italy
| | - Aurora García-Tejedor
- Bioactivity and Nutritional Immunology Group (BIOINUT), Faculty of Health Sciences, Universidad Internacional de Valencia-VIU, Pintor Sorolla 21, 46002 Valencia, Spain
| | - Claudia Monika Haros
- Instituto de Agroquímica y Tecnología de Alimentos (IATA), Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain
| | - José Moisés Laparra Llopis
- Molecular Immunonutrition Group, Madrid Institute for Advanced Studies in Food (IMDEA-Food), Madrid, Spain.
| |
Collapse
|
3
|
A Three-Gene Signature for Predicting the Prognosis of Patients Treated with Transarterial Chemoembolization (TACE) and Identification of PD-184352 as a Potential Drug to Reverse Nonresponse to TACE. JOURNAL OF ONCOLOGY 2022; 2022:2704862. [PMID: 36213835 PMCID: PMC9534656 DOI: 10.1155/2022/2704862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/08/2022] [Accepted: 07/31/2022] [Indexed: 11/30/2022]
Abstract
Background Transarterial chemoembolization (TACE) is a first-line treatment for patients with unresectable hepatocellular carcinoma (HCC). Owing to differences in its efficacy across individuals, determining the indicators of patient response to TACE and finding approaches to reversing nonresponse thereto are necessary. Methods Transcriptome data were obtained from the GSE104580 dataset, in which patients were marked as having TACE response or nonresponse. We identified differentially expressed genes (DEGs) and performed Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. We screened genes with a prognostic value for TACE in the HIF-1 signaling pathway by univariate regression analysis. By using least absolute shrinkage and selection operator (LASSO) Cox regression, we established a multigene signature in GSE14520, which we verified using a drug sensitivity test. The Connectivity Map (CMap) database was used to find potential drugs to reverse nonresponse to TACE. Results We constructed a prognostic signature consisting of three genes (erythropoietin (EPO), heme oxygenase 1 (HMOX1), and serine protease inhibitor 1 (SERPINE1)) that we validated by drug sensitivity test. After dividing patients treated with TACE into high- and low-risk groups based on this new signature, we showed that overall survival (OS) of the high-risk group was significantly lower than that of the low-risk group and that the risk score was an independent predictor of OS in patients treated with TACE. Based on our CMap findings, we speculated that PD-184352, an inhibitor of mitogen-activated protein kinase (MEK), had potential as a drug treatment to reverse nonresponse to TACE. We confirmed this speculation by using PD-184352 in a cell promotion experiment in a TACE environment. Conclusion We constructed a TACE-specific three-gene signature that could be used to predict HCC patients' responses to and prognosis after TACE treatment. PD-184352 might have potential as a drug to improve TACE efficacy.
Collapse
|
4
|
Expression Dynamics of Heme Oxygenase-1 in Tumor Cells and the Host Contributes to the Progression of Tumors. J Pers Med 2021; 11:jpm11121340. [PMID: 34945812 PMCID: PMC8704574 DOI: 10.3390/jpm11121340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 01/05/2023] Open
Abstract
Heme oxygenase (HO-1) plays an important role in cellular protection against various stresses. The induction of HO-1 is an effective strategy for reactive oxygen species-related diseases, inflammatory diseases, as well as suppressing carcinogenesis. On the other hand, the high expression of HO-1 is now well known in many tumors. In this study, we investigated the dynamics of HO-1 expression in the host and the tumor. In the mouse sarcoma S180 solid tumor model and the rat hepatoma AH136B ascitic tumor model, HO-1 expression in the tumor, as indicated by the end product of HO-1 activation, i.e., carbon monoxide, gradually increased along with tumor growth. Over-expression of HO-1 expression in mouse colon cancer C26 tumor cells significantly promoted tumor growth as well as lung metastasis, whereas opposite results were found when the HO-1 expression was reduced in the cells. On the other hand, upregulating HO-1 levels in the host by using an HO-1 inducer protected the progression of the xenograft tumor in mice, whereas lowering HO-1 expression in the host with an HO-1 inhibitor showed accelerated tumor growth and lung metastasis after subcutaneous tumor xenograft inoculation. These findings strongly suggest that the balance of HO-1 levels in the host and the tumor cells is essential for the occurrence, progression, and prognosis of cancer. Maintenance of appropriately high HO-1 levels in the host is favorable for cancer prevention, whereas suppression of HO-1 in the tumor cells may thus become a therapeutic strategy for cancer.
Collapse
|
5
|
Abstract
Significance: As the central metabolic organ, the liver is exposed to a variety of potentially cytotoxic, proinflammatory, profibrotic, and carcinogenic stimuli. To protect the organism from these deleterious effects, the liver has evolved a number of defense systems, which include antioxidant substrates and enzymes, anti-inflammatory tools, enzymatic biotransformation systems, and metabolic pathways. Recent Advances: One of the pivotal systems that evolved during phylogenesis was the heme catabolic pathway. Comprising the important enzymes heme oxygenase and biliverdin reductase, this complex pathway has a number of key functions including enzymatic activities, but also cell signaling, and DNA transcription. It further generates two important bile pigments, biliverdin and bilirubin, as well as the gaseous molecule carbon monoxide. These heme degradation products have potent antioxidant, immunosuppressive, and cytoprotective effects. Recent data suggest that the pathway participates in the regulation of metabolic and hormonal processes implicated in the pathogenesis of hepatic and other diseases. Critical Issues: This review discusses the impact of the heme catabolic pathway on major liver diseases, with particular focus on the involvement of cellular targeting and signaling in the pathogenesis of these conditions. Future Directions: To utilize the biological consequences of the heme catabolic pathway, several unique therapeutic strategies have been developed. Research indicates that pharmaceutical, nutraceutical, and lifestyle modifications positively affect the pathway, delivering potentially long-term clinical benefits. However, further well-designed studies are needed to confirm the clinical benefits of these approaches. Antioxid. Redox Signal. 35, 734-752.
Collapse
Affiliation(s)
- Libor Vítek
- Fourth Department of Internal Medicine, and Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
6
|
Wei J, Ma L, Liu W, Wang Y, Shen C, Zhao X, Zhao C. Identification of the molecular targets and mechanisms of compound mylabris capsules for hepatocellular carcinoma treatment through network pharmacology and bioinformatics analysis. JOURNAL OF ETHNOPHARMACOLOGY 2021; 276:114174. [PMID: 33932512 DOI: 10.1016/j.jep.2021.114174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/21/2021] [Accepted: 04/24/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese herbal formulas have been proven to exert an inhibitory effect on tumor. Compound mylabris capsules (CMC) has been used for treating cancer, especially hepatocellular carcinoma (HCC), for years in China. However, its therapeutic mechanisms on HCC remain unclear. AIM OF THE STUDY This research aimed to elucidate the molecular targets and mechanisms of CMC for treating HCC. MATERIALS AND METHODS First, the bioactive ingredients and potential targets of CMC, as well as HCC-related targets were retrieved from publicly available databases. Next, the overlapped genes between potential targets of CMC and HCC-related targets were determined using bioinformatics analysis. Then, networks of ingredient-target and gene-pathway were constructed. Finally, cell experiments were carried out to examine the effects of CMC-medicated serum on HCC and validate the core molecular targets. RESULTS In total, 151 bioactive ingredients and 255 potential targets of CMC were selected, 982 differentially expressed genes of HCC were identified. Among them, 34 overlapped genes were finally selected. In addition, 20 pathways and 429 GO terms were significantly enriched. Protein-protein interaction and gene-pathway networks indicated that Cyclin B1(CCNB1) and Cyclin Dependent Kinase 1(CDK1) were the core gene targets for the treatment of CMC on HCC. Moreover, in vitro studies showed that CMC-medicated serum significantly inhibited the viability of HepG2 cells. Furthermore, CMC downregulated CCNB1 and CDK1 expressions and induced G2/M phase cell cycle arrest. CONCLUSIONS CMC plays a therapeutic role in HCC via multi-component, -target and -pathway mechanisms, in which CCNB1 and CDK1 may be the core molecular targets. This study indicates that the integration of network pharmacology and bioinformatics analysis, followed by experimental validation, can serves as an effective tool for studying the therapeutic mechanisms of traditional Chinese medicine.
Collapse
Affiliation(s)
- Junwei Wei
- Department of Infectious Diseases, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China.
| | - Luyuan Ma
- Department of Infectious Diseases, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China.
| | - Wenpeng Liu
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China.
| | - Yadong Wang
- Department of Infectious Diseases, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China.
| | - Chuan Shen
- Department of Infectious Diseases, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China.
| | - Xin Zhao
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China.
| | - Caiyan Zhao
- Department of Infectious Diseases, Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China.
| |
Collapse
|
7
|
GT-Repeat Polymorphism in the HO-1 Gene Promoter Is Associated with Risk of Liver Cancer: A Follow-Up Study from Arseniasis-Endemic Areas in Taiwan. J Clin Med 2021; 10:jcm10071489. [PMID: 33916685 PMCID: PMC8038349 DOI: 10.3390/jcm10071489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 11/16/2022] Open
Abstract
The induction of heme oxygenase-1 (HO-1) has been shown to have therapeutic potential in experimental models of hepatitis and liver fibrosis, which are closely related to liver cancer. In humans, HO-1 induction is transcriptionally modulated by the length of a GT-repeat [(GT)n] in the promoter region. We aimed to investigate the effect of HO-1 (GT)n variants on liver cancer in a human population. We determined the HO-1 genotype in 1153 study subjects and examined their association with liver cancer risk during a 15.9-year follow-up. Allelic polymorphisms were classified as short [S, <27 (GT)n] or long [L, ≥27 (GT)n]. Newly developed cancer cases were identified through linkage to the National Cancer Registry of Taiwan. Multivariate Cox regression analysis was used to evaluate the effect of the HO-1 (GT)n variants. Alpha-fetoprotein (AFP) and cirrhosis history were also examined. The S/S genotype was found to be significantly associated with liver cancer risk, compared to the L/S and L/L genotypes. The S/S genotype group also had a higher percentage of subjects with abnormal AFP levels than other groups. There were significant percentages of cirrhosis among groups who carried S-alleles. Our findings indicate that short (GT)n variants in the HO-1 gene may confer susceptibility to rather than protection from liver cirrhosis/cancer.
Collapse
|
8
|
Li R, Zhang J, Zhou Y, Gao Q, Wang R, Fu Y, Zheng L, Yu H. Transcriptome Investigation and In Vitro Verification of Curcumin-Induced HO-1 as a Feature of Ferroptosis in Breast Cancer Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3469840. [PMID: 33294119 PMCID: PMC7691002 DOI: 10.1155/2020/3469840] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/07/2020] [Accepted: 10/29/2020] [Indexed: 12/24/2022]
Abstract
Ferroptosis is a form of oxidative cell death and has become a chemotherapeutic target for cancer treatment. Curcumin (CUR), a well-known cancer inhibitor, significantly inhibits the viability of breast cancer cells. Through transcriptomic analysis and flow cytometry experiments, it was found that after 48 hours of treatment of breast cancer cells at its half maximal inhibitory concentration (IC50), curcumin suppressed the viability of cancer cells via induction of ferroptotic death. Use of the ferroptosis inhibitor ferrostatin-1 and the iron chelator deferoxamine rescued cell death induced by curcumin. Furthermore, in subsequent cell validation experiments, the results showed that curcumin caused marked accumulation of intracellular iron, reactive oxygen species, lipid peroxides, and malondialdehyde, while glutathione levels were significantly downregulated. These changes are all manifestations of ferroptosis. Curcumin upregulates a variety of ferroptosis target genes related to redox regulation, especially heme oxygenase-1 (HO-1). Using the specific inhibitor zinc protoporphyrin 9 (ZnPP) to confirm the above experimental results showed that compared to the curcumin treatment group, treatment with ZnPP not only significantly improved cell viability but also reduced the accumulation of intracellular iron ions and other ferroptosis-related phenomena. Therefore, these data demonstrate that curcumin triggers the molecular and cytological characteristics of ferroptosis in breast cancer cells, and HO-1 promotes curcumin-induced ferroptosis.
Collapse
Affiliation(s)
- Ruihua Li
- College of Animal Science, Jilin University, Changchun 130062, China
| | - Jing Zhang
- College of Animal Science, Jilin University, Changchun 130062, China
- Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China
| | - Yongfeng Zhou
- College of Animal Science, Jilin University, Changchun 130062, China
| | - Qi Gao
- Biological Emergency and Clinical POCT Key Laboratory, Beijing 102600, China
| | - Rui Wang
- College of Animal Science, Jilin University, Changchun 130062, China
| | - Yurong Fu
- College of Animal Science, Jilin University, Changchun 130062, China
| | - Lianwen Zheng
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun 130022, China
| | - Hao Yu
- College of Animal Science, Jilin University, Changchun 130062, China
| |
Collapse
|
9
|
Rossier J, Delasoie J, Haeni L, Hauser D, Rothen-Rutishauser B, Zobi F. Cytotoxicity of Mn-based photoCORMs of ethynyl-α-diimine ligands against different cancer cell lines: The key role of CO-depleted metal fragments. J Inorg Biochem 2020; 209:111122. [PMID: 32497818 DOI: 10.1016/j.jinorgbio.2020.111122] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 12/19/2022]
Abstract
A series of tricarbonyl manganese complexes bearing 4-ethynyl-2,2'-bipyridine and 5-ethynyl-1,10-phenanthroline α-diimine ligands were synthetized, characterized and conjugated to vitamin B12, previously used as a vector for drug delivery, to take advantage of its water solubility and specificity toward cancer cells. The compounds act as photoactivatable carbon monoxide-releasing molecules rapidly liberating on average ca. 2.3 equivalents of CO upon photo-irradiation. Complexes and conjugates were tested for their anticancer effects, both in the dark and following photo-activation, against breast cancer MCF-7, lung carcinoma A549 and colon adenocarcinoma HT29 cell lines as well as immortalized human bronchial epithelial cells 16HBE14o- as the non-carcinogenic control. Our results indicate that the light-induced cytotoxicity these molecules can be attributed to both their released CO and to their CO-depleted metal fragments including liberated ligands.
Collapse
Affiliation(s)
- Jeremie Rossier
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland
| | - Joachim Delasoie
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland
| | - Laetitia Haeni
- Adolphe Merkle Institute, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | - Daniel Hauser
- Adolphe Merkle Institute, Chemin des Verdiers 4, 1700 Fribourg, Switzerland
| | | | - Fabio Zobi
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland.
| |
Collapse
|
10
|
Chiang KC, Chang KS, Hsu SY, Sung HC, Feng TH, Chao M, Juang HH. Human Heme Oxygenase-1 Induced by Interleukin-6 via JAK/STAT3 Pathways Is a Tumor Suppressor Gene in Hepatoma Cells. Antioxidants (Basel) 2020; 9:antiox9030251. [PMID: 32204510 PMCID: PMC7139670 DOI: 10.3390/antiox9030251] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 12/13/2022] Open
Abstract
Heme oxygenase-1 (HO-1) has several important roles in hepatocytes in terms of anti-inflammation, anti-apoptosis, and antioxidant properties. Interleukin-6 (IL-6) is a pleiotropic cytokine associated with liver regeneration and protection against injury. The aim of this study was to determine the potential crosstalk between HO-1 and IL-6, and to elucidate the signaling pathways involved in the induction of HO-1 by IL-6 in human hepatoma cells. Ectopic overexpression of HO-1 not only attenuated cell proliferation in vitro and in vivo, but also blocked the reactive oxygen species (ROS) induced by H2O2 and the pyocyanin in HepG2 or Hep3B cells. IL-6 expression was negatively regulated by HO-1, while IL-6 induced signal transducer and activator of transcription 3 (STAT3) phosphorylation and HO-1 gene expression in HepG2 cells. The co-transfected HO-1 reporter vector and a protein inhibitor of the activated STAT3 (PIAS3) expression vector blocked the IL-6-induced HO-1 reporter activity. Both interferon γ and interleukin-1β treatments induced STAT1 but not STAT3 phosphorylation, which had no effects on the HO-1 expression. Treatments of AG490 and luteolin blocked the JAK/STAT3 signaling pathways which attenuated IL-6 activation on the HO-1 expression. Our results indicated that HO-1 is the antitumor gene induced by IL-6 through the IL-6/JAK/STAT3 pathways; moreover, a feedback circuit may exist between IL-6 and HO-1 in hepatoma cells.
Collapse
Affiliation(s)
- Kun-Chun Chiang
- Department of General Surgery, Min-Sheng General Hospital, Tao-Yuan 33302, Taiwan;
| | - Kang-Shuo Chang
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-S.C.); (S.-Y.H.); (H.-C.S.)
- Institute of Medicine Science, College of Medicine, ChSang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan
| | - Shu-Yuan Hsu
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-S.C.); (S.-Y.H.); (H.-C.S.)
| | - Hsin-Ching Sung
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-S.C.); (S.-Y.H.); (H.-C.S.)
| | - Tsui-Hsia Feng
- School of Nursing, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan;
| | - Mei Chao
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan
- Department of Hepato-Gastroenterology, Liver Research Center, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Tao-Yuan 33302, Taiwan
- Correspondence: (M.C.); (H.-H.J.); Tel.:+886-3-2118800 (M.C. & H.-H.J.); Fax: +886-3-2118112 (M.C. & H.-H.J.)
| | - Horng-Heng Juang
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-S.C.); (S.-Y.H.); (H.-C.S.)
- Institute of Medicine Science, College of Medicine, ChSang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Tao-Yuan 33302, Taiwan
- Correspondence: (M.C.); (H.-H.J.); Tel.:+886-3-2118800 (M.C. & H.-H.J.); Fax: +886-3-2118112 (M.C. & H.-H.J.)
| |
Collapse
|
11
|
Canesin G, Hejazi SM, Swanson KD, Wegiel B. Heme-Derived Metabolic Signals Dictate Immune Responses. Front Immunol 2020; 11:66. [PMID: 32082323 PMCID: PMC7005208 DOI: 10.3389/fimmu.2020.00066] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/10/2020] [Indexed: 12/21/2022] Open
Abstract
Heme is one of the most abundant molecules in the body acting as the functional core of hemoglobin/myoglobin involved in the O2/CO2 carrying in the blood and tissues, redox enzymes and cytochromes in mitochondria. However, free heme is toxic and therefore its removal is a significant priority for the host. Heme is a well-established danger-associated molecular pattern (DAMP), which binds to toll-like receptor 4 (TLR4) to induce immune responses. Heme-derived metabolites including the bile pigments, biliverdin (BV) and bilirubin (BR), were first identified as toxic drivers of neonatal jaundice in 1800 but have only recently been appreciated as endogenous drivers of multiple signaling pathways involved in protection from oxidative stress and regulators of immune responses. The tissue concentration of heme, BV and BR is tightly controlled. Heme oxygenase-1 (HO-1, encoded by HMOX1) produces BV by heme degradation, while biliverdin reductase-A (BLVR-A) generates BR by the subsequent conversion of BV. BLVR-A is a fascinating protein that possesses a classical protein kinase domain, which is activated in response to BV binding to its enzymatic site and initiates the downstream mitogen-activated protein kinases (MAPK) and phosphatidylinositol 3-kinase (PI3K) pathways. This links BLVR-A activity to cell growth and survival pathways. BLVR-A also contains a bZip DNA binding domain and a nuclear export sequence (NES) and acts as a transcription factor to regulate the expression of immune modulatory genes. Here we will discuss the role of heme-related immune response and the potential for targeting the heme system for therapies directed toward hepatitis and cancer.
Collapse
Affiliation(s)
- Giacomo Canesin
- Department of Surgery, Cancer Research Institute and Transplant Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Seyed M. Hejazi
- Department of Surgery, Cancer Research Institute and Transplant Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Kenneth D. Swanson
- Brain Tumor Center and Neuro-Oncology Unit, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Barbara Wegiel
- Department of Surgery, Cancer Research Institute and Transplant Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
12
|
Abstract
Gas-involving cancer theranostics have attracted considerable attention in recent years due to their high therapeutic efficacy and biosafety. We have reviewed the recent significant advances in the development of stimuli-responsive gas releasing molecules (GRMs) and gas nanogenerators for cancer bioimaging, targeted and controlled gas therapy, and gas-sensitized synergistic therapy. We have focused on gases with known anticancer effects, such as oxygen (O2), carbon monoxide (CO), nitric oxide (NO), hydrogen sulfide (H2S), hydrogen (H2), sulfur dioxide (SO2), carbon dioxide (CO2), and heavy gases that act via the gas-generating process. The GRMs and gas nanogenerators for each gas have been described in terms of the stimulation method, followed by their applications in ultrasound and multimodal imaging, and finally their primary and synergistic actions with other cancer therapeutic modalities. The current challenges and future possibilities of gas therapy and imaging vis-à-vis clinical translation have also been discussed.
Collapse
Affiliation(s)
- Lichan Chen
- College of Chemical Engineering , Huaqiao University , Xiamen , Fujian 361021 , P.R. China
| | - Shu-Feng Zhou
- College of Chemical Engineering , Huaqiao University , Xiamen , Fujian 361021 , P.R. China
| | - Lichao Su
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry , Fuzhou University , Fuzhou , Fujian 350116 , P.R. China
| | - Jibin Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry , Fuzhou University , Fuzhou , Fujian 350116 , P.R. China
| |
Collapse
|
13
|
Li Q, Liu Q, Cheng W, Wei H, Jiang W, E F, Yu Y, Jin J, Zou C. Heme Oxygenase-1 Inhibits Tumor Metastasis Mediated by Notch1 Pathway in Murine Mammary Carcinoma. Oncol Res 2019; 27:643-651. [PMID: 30764900 PMCID: PMC7848234 DOI: 10.3727/096504018x15415906335771] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Heme oxygenase-1 (HO-1) plays an important role in the progression of several malignancies including breast cancer. However, its role in breast cancer metastasis is still ambiguous. In this study, we observed the effect of HO-1 on mouse mammary carcinoma metastasis using the in vivo tumor metastasis model. Our results revealed that overexpression of HO-1 strongly inhibits the lung metastasis of 4T1 cells. In in vitro analysis, associated indices for epithelial-mesenchymal transition (EMT), migration, and proliferation of 4T1 cells were evaluated. The results show that HO-1 inhibits EMT, migration, and proliferation of 4T1 cells. In addition, the Notch1/Slug pathway is found to mediate an antimetastasis role of HO-1 in mouse mammary carcinoma. In conclusion, since HO-1/Notch1/Slug axis plays an important role in breast cancer metastasis, induction of HO-1 could be used as a potential therapeutic strategy for breast cancer treatment.
Collapse
Affiliation(s)
- Qiang Li
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
- †Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, P.R. China
| | - Qi Liu
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
| | - Wanpeng Cheng
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
| | - Huiyan Wei
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
| | - Wenqian Jiang
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
| | - Fang E
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
| | - Yuan Yu
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
| | - Jianfeng Jin
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
| | - Chaoxia Zou
- *Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, P.R. China
- ‡Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medicine Sciences, Harbin, P.R. China
| |
Collapse
|
14
|
Park CS, Eom DW, Ahn Y, Jang HJ, Hwang S, Lee SG. Can heme oxygenase-1 be a prognostic factor in patients with hepatocellular carcinoma? Medicine (Baltimore) 2019; 98:e16084. [PMID: 31261522 PMCID: PMC6617477 DOI: 10.1097/md.0000000000016084] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is an important catalytic enzyme in heme degradation, which increases during stressful conditions. It plays a major role in antioxidative and antiapoptotic processes and is associated with tumor growth and metastasis.This study aimed to evaluate the degree of HO-1 expressions in hepatocellular carcinoma (HCC) surgical specimens and the correlation between HO-1 expression and patient prognosis. Formalin-fixed, paraffin-embedded HCC tissue samples (n = 96) were included in the analysis, and the expression of HO-1 was evaluated by immunohistochemical staining. We reviewed clinical features of patients and evaluated the prognostic role of HO-1 in patient survival and recurrence.Positive HO-1 expression was identified in 43 cases (44.8%) and was frequently found in patients with advanced histology (Edmondson-Steiner [E-S] grade 2, 3, 4), α-fetoprotein (AFP) level of more than 200 IU/mL, and the presence of microvascular and capsular invasion (P < .05). In the univariate analysis, the overall survival (OS) and disease-free survival (DFS) of patients with HO-1-positive HCC were not statistically different from those with HO-1-negative HCC. Moreover, HO-1 expression was not associated with patient survival and recurrence based on the multivariate analysis. In the subgroup analysis of patients without preoperative transarterial chemoembolization (TACE) (n = 61), HO-1 was not also associated with tumor recurrence (P = .681).The clinical implication of HO-1 activity is controversial in various malignancies. However, HO-1 expression did not seem to influence the prognosis of HCC patients.
Collapse
Affiliation(s)
- Cheon-Soo Park
- Department of Surgery, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea
| | - Dae-Woon Eom
- Department of Pathology, Gangneung Asan Hospital, University of Ulsan College of Medicine
| | - Yongchel Ahn
- Department of Hematology and Oncology, Gangneung Asan Hospital, University of Ulsan College of Medicine
| | - Hyuk Jai Jang
- Department of Surgery, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung
| | - Shin Hwang
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Gyu Lee
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
15
|
Li S, Li G, Zhang T, Li J, Zhao Q, Zhang B, Wang R, Zhou R, Si J, Gan L, Liu Y, Zhang H, Liu B. Co-SLD suppressed the growth of oral squamous cell carcinoma via disrupting mitochondrial function. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:1746-1757. [PMID: 31062618 DOI: 10.1080/21691401.2019.1608218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Sirui Li
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Guo Li
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Taofeng Zhang
- School of Nuclear Science and Technology, Lanzhou University, Lanzhou, China
| | - Jili Li
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Quanyi Zhao
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Baoping Zhang
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Rui Wang
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Rong Zhou
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Jing Si
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Lu Gan
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Yang Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Hong Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Bin Liu
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, China
| |
Collapse
|
16
|
Kim HN, Park GH, Park SB, Kim JD, Eo HJ, Son HJ, Song JH, Jeong JB. Extracts from Sageretia thea reduce cell viability through inducing cyclin D1 proteasomal degradation and HO-1 expression in human colorectal cancer cells. Altern Ther Health Med 2019; 19:43. [PMID: 30736789 PMCID: PMC6368743 DOI: 10.1186/s12906-019-2453-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/30/2019] [Indexed: 12/12/2022]
Abstract
Background Sageretia thea (S. thea) has been used as the medicinal plant for treating hepatitis and fevers in Korea and China. Recently, anticancer activity of S. thea has been reported, but the potential mechanism for the anti-cancer property of S. thea is still insufficient. Thus, we evaluated whether extracts from the leaves (STL) and branches (STB) of S. thea exert anticancer activity and elucidated its potential mechanism in SW480 cells. Methods MTT assay was performed for measuring cell viability. Western blot and RT-PCR were used for analyzing the level of protein and mRNA, respectively. Results Treatment of STL or STB decreased the cell viability and induced apoptosis in SW480 cells. Decreased level of cyclin D1 protein was observed in SW480 cells treated with STL or STB, but no change in cyclin D1 mRNA level was observed with the treatment of STL or STB. MG132 blocked downregulation of cyclin D1 protein by STL or STB. Thr286 phosphorylation of cyclin D1 by STL or STB occurred faster than downregulation of cyclin D1 protein in SW480 cells. When SW480 cells were transfected with T286A-cyclin D1, cyclin D1 degradation by STL or STB did not occur. Inhibition of GSK3β and cyclin D1 nuclear export attenuated STL or STB-mediated cyclin D1 degradation. In addition, STL or STB increased HO-1 expression, and the inhibition of HO-1 attenuated the induction of apoptosis by STL or STB. HO-1 expression by STL or STB resulted from Nrf2 activation through ROS-dependent p38 activation. Conclusions These results indicate that STL or STB may induce GSK3β-dependent cyclin D1 degradation, and increase HO-1 expression through activating Nrf2 via ROS-dependent p38 activation, which resulted in the decrease of the viability in SW480 cells. These findings suggest that STL or STB may have great potential for the development of anti-cancer drug.
Collapse
|
17
|
Kourti M, Westwell A, Jiang W, Cai J. Repurposing old carbon monoxide-releasing molecules towards the anti-angiogenic therapy of triple-negative breast cancer. Oncotarget 2019; 10:1132-1148. [PMID: 30800223 PMCID: PMC6383690 DOI: 10.18632/oncotarget.26638] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 01/16/2019] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is defined by the lack of expression of the oestrogen and progesterone receptors and HER-2. Recently, carbon monoxide (CO) was found to behave as an important endogenous signalling molecule and to suppress VEGF receptor-2 (VEGFR-2) and protein kinase B phosphorylation. Given that anti-angiogenic drugs exist as one of the few available targeted therapies against TNBC, the aim of this project was to study the effects of CO-releasing molecules (CORMs) on TNBC cell lines and the associated endothelial cells and characterise their anti-angiogenic properties that can be used for the reduction of cancer-driven angiogenesis. Four commercially available CORMs were screened for their cytotoxicity, their effects on cell metabolism, migration, VEGF expression, tube formation and VEGFR-2 activation. The most important result was the reduction in VEGF levels expressed by CORM-treated TNBC cells, along with the inhibition of phosphorylation of VEGFR2 and downstream proteins. The migration and tube formation ability of endothelial cells was also decreased by CORMs, justifying a potential re-purposing of old CORMs towards the anti-angiogenic therapy of TNBC. The additional favourable low cytotoxicity, reduction in the glycolysis levels and downregulation of haem oxygenase-1 in TNBC cells enhance the potential of CORMs against TNBC. In this study, CORM-2 remained the most effective CORM and we propose that CORM-2 may be pursued further as an additional agent in combination with existing anti-angiogenic therapies for a more successful targeting of malignant angiogenesis in TNBC.
Collapse
Affiliation(s)
- Malamati Kourti
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK.,School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, CF10 3NB, UK
| | - Andrew Westwell
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, CF10 3NB, UK
| | - Wen Jiang
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Jun Cai
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| |
Collapse
|
18
|
Yang Y, Wang HX, Zhang L, Huo W, Li XD, Qi RQ, Song XY, Wei S, Gao XH, Han S, Cao L. Inhibition of Heme Oxygenase-1 enhances hyperthermia-induced autophagy and antiviral effect. Int J Biol Sci 2019; 15:568-578. [PMID: 30745843 PMCID: PMC6367586 DOI: 10.7150/ijbs.29759] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/15/2018] [Indexed: 12/19/2022] Open
Abstract
Hyperthermia has been clinically utilized as an adjuvant therapy in the treatment of cervical carcinoma. However, thermotolerance induced by heme oxygenase-1 (HO-1), a stress-inducible cytoprotective protein, limits the efficacy of hyperthermic therapy, for which the exact mechanism remains unknown. In the present study, we found that heat treatment induced HO-1 expression and decreased copy number of HPV16 in cervical cancer cells and tissues from cervical cancer and precursor lesions. Knockdown of HO-1 stimulated autophagy accompanied by downregulation of X-linked inhibitor of apoptosis protein. Furthermore, silencing of HO-1 led to cell intolerance to hyperthermia, as manifested by inhibition of cell viability and induction of autophagic apoptosis. Moreover, HO-1 modulated hyperthermia-induced, autophagy-dependent antiviral effect. Thus, the findings indicate that blockade of HO-1 enhances hyperthermia-induced autophagy, an event resulting in apoptosis of cervical cancer cells through an antiviral mechanism. These observations imply the potential clinical utility of hyperthermia in combination with HO-1 inhibition in the treatment of cervical cancer.
Collapse
Affiliation(s)
- Yang Yang
- Department of Dermatology, No.1 Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang 110001, China.,Key Laboratory of Medical Cell Biology, China Medical University, Shenyang, 110122, China
| | - He-Xiao Wang
- Department of Dermatology, No.1 Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang 110001, China
| | - Lan Zhang
- Department of Dermatology, No.1 Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang 110001, China
| | - Wei Huo
- Department of Dermatology, No.1 Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang 110001, China
| | - Xiao-Dong Li
- Department of Dermatology, Central Hospital Affiliated to Shen Yang Medical College, Shenyang, 110001, China
| | - Rui-Qun Qi
- Department of Dermatology, No.1 Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang 110001, China
| | - Xiao-Yu Song
- Key Laboratory of Medical Cell Biology, China Medical University, Shenyang, 110122, China
| | - Shi Wei
- Department of Pathology, the University of Alabama at Birmingham, Birmingham, Alabama 35249, United States
| | - Xing-Hua Gao
- Department of Dermatology, No.1 Hospital of China Medical University and Key Laboratory of Immunodermatology, Ministry of Health and Ministry of Education, Shenyang 110001, China
| | - Shuai Han
- Department of Neurosurgery, No.1 Hospital of China Medical University, Shenyang 110001, China
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, China Medical University, Shenyang, 110122, China
| |
Collapse
|
19
|
Yeh CN, Wu RC, Cheng CT, Tsai CY, Chang YR, Yeh TS, Wu TH, Lee WC, Chiang KC. HO-1 is a favorable prognostic factor for HBV-HCC patients who underwent hepatectomy. Cancer Manag Res 2018; 10:6049-6059. [PMID: 30538554 PMCID: PMC6252782 DOI: 10.2147/cmar.s186931] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background More than 500,000 people suffered from hepatocelluar carcinoma (HCC) annually and the relative incidence to mortality rate indicates its unfavorable prognosis. Several studies have proved that heme-oxygenase-1 (HO-1) is indirectly engaged in the invasion and the metastasis of some types of malignancies, including breast cancer, prostate cancer, and lung cancer. The role of HO-1 in hepatitis B virus (HBV)-related HCC is still not clarified. Materials and methods The Western blot, doubling time, cell cycle analysis, migration assay, invasion assay, gene transfection, xenograft animal model, immunohistochemistry staining, and clinical validation study were applied in this study. Results HO-1 overexpression not only decreased the growth but also inhibited the migration and invasion in human HBV-HCC cells (Hep-3B vs PLC/PRF/5). The inhibitory effect on growth, migration, and invasion is further demonstrated by the overexpression of HO-1 in Hep-3B cell by transfection study. Furthermore, HO-1 decreasing the growth of HBV-HCC was confirmed in animal study. The clinical validation illustrated that higher HO1 expression was also associated with favorable disease-free survival of HBV-HCC patients who underwent hepatectomy. Conclusions We identified HO-1 as a favorable prognostic factor for HBV-HCC patients who underwent hepatectomy.
Collapse
Affiliation(s)
- Chun-Nan Yeh
- Department of Surgery, Liver Research Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan,
| | - Ren-Chin Wu
- Department of Pathology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan
| | - Chi-Tung Cheng
- Department of Surgery, Liver Research Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan,
| | - Chun-Yi Tsai
- Department of Surgery, Liver Research Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan,
| | - Yau-Ren Chang
- Department of Surgery, Liver Research Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan,
| | - Ta-Sen Yeh
- Department of Surgery, Liver Research Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan,
| | - Tsung-Han Wu
- Department of Surgery, Liver Research Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan,
| | - Wei-Chen Lee
- Department of Surgery, Liver Research Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan,
| | - Kun-Chun Chiang
- General Surgery Department, Chang Gung Memorial Hospital, Chang Gung University, Keelung, Taiwan,
| |
Collapse
|
20
|
Huang H, Li Y, Wu M, Luo J, Nie J, Hou B, He Q, Diao Y, Qi L, Zhao Y, Liu Y, Yang D, Zhou L. Effects of ethanol on the anticancer function of doxorubicin in JJ012 cells. Future Oncol 2018; 14:1285-1297. [PMID: 29774752 DOI: 10.2217/fon-2017-0547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Chondrosarcoma is difficult to treat because of resistance to conventional chemotherapy and radiotherapy. This study evaluated the effects of ethanol in combination with doxorubicin in chondrosarcoma cells. MATERIALS & METHODS JJ012, was treated with doxorubicin alone or in combination with ethanol. Effects on cellular proliferation, migration, invasion, apoptosis, and the cell cycle were evaluated. RESULTS Treatment of JJ012 cells with 100 mM ethanol and doxorubicin resulted in reduced cell growth, invasion, and migration. In addition, doxorubicin uptake into the nucleus was enhanced and p53 mRNA expression was upregulated in JJ012 cells. CONCLUSION Ethanol combined with doxorubicin increased doxorubicin uptake in the nucleus and enhanced the effects of doxorubicin in JJ012 cells.
Collapse
Affiliation(s)
- Hui Huang
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China.,Basic Medical Institute of Heilongjiang Medical Science Academy, Harbin, PR China.,Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, PR China
| | - Yanze Li
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China.,Basic Medical Institute of Heilongjiang Medical Science Academy, Harbin, PR China.,Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, PR China
| | - Mingjuan Wu
- Heilongjiang Academy of Traditional Chinese Medicine, Harbin, PR China
| | - Jing Luo
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China
| | - Junhui Nie
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China
| | - Baoyu Hou
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China
| | - Qi He
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China
| | - Yan Diao
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China
| | - Lin Qi
- Department of Radioimmunossay, Heilongjiang Province Hospital, Harbin, PR China
| | - Yuanyuan Zhao
- Department of Anesthesiology, Heilongjiang Province Hospital, Harbin, PR China
| | - Ying Liu
- Department of Gastroenterology, Heilongjiang Province Hospital, Harbin, PR China
| | - Dan Yang
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China.,Basic Medical Institute of Heilongjiang Medical Science Academy, Harbin, PR China.,Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, PR China
| | - Lingyun Zhou
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, PR China.,Translational Medicine Center of Northern China, Harbin Medical University, Harbin, PR China.,Basic Medical Institute of Heilongjiang Medical Science Academy, Harbin, PR China.,Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, PR China
| |
Collapse
|
21
|
Wójcik M, Bobowiec R, Lisiecka U, Śmiech A. Expression of receptor interacting protein 1 and receptor interacting protein 3 oval cells in a rat model of hepatocarcinogenesis. Exp Ther Med 2018; 15:4448-4456. [PMID: 29731829 DOI: 10.3892/etm.2018.5988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 02/08/2018] [Indexed: 01/25/2023] Open
Abstract
When apoptosis is suppressed in a neoplastic state, necroptosis may enable an anticancer response. In the present study, the association between apoptosis and necroptosis was assessed in a partial hepatectomy (PH)/diethylnitrosamine (DEN) rat model of hepatocarcinogenesis. Isolated oval cells (OCs) were analysed at 24, 48 and 72 h and at the first and second week of incubation. Phenotypic studies, apoptosis and necroptosis detection and proliferative activity assays were also performed on the OCs. The OCs were isolated from non-neoplastic (PH) and neoplastic (PH/DEN) livers, which expressed receptor interacting protein (RIP) 1 and RIP3. Western blot analysis revealed that the RIP1 and RIP3 expression in the PH/DEN OCs started to increase at 72 h and continually increased to the end of cell culture. Compared with the PH OCs, the cells isolated from PH/DEN rats exhibited significantly less potential for apoptosis (P<0.05). There were a minimal number of apoptotic PH/DEN OCs (2.82±1.1%) at 72 h. In addition, the PH/DEN OCs demonstrated progressive proliferative activity during incubation, which was significantly increased compared with the PH OCs at ≥72 h. The present study revealed that PH/DEN OCs, which trigger hepatic cancer, have a high proliferative activity and suppress apoptosis. It was also observed that, based on the expression of RIP3 and RIP1, necroptosis may be maintained and may serve as an alternative pathway for programmed PH/DEN OC death.
Collapse
Affiliation(s)
- Marta Wójcik
- Department of Pathophysiology, Faculty of Veterinary Medicine, University of Life Sciences, 20-033 Lublin, Poland
| | - Ryszard Bobowiec
- Department of Pathophysiology, Faculty of Veterinary Medicine, University of Life Sciences, 20-033 Lublin, Poland
| | - Urszula Lisiecka
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences, 20-033 Lublin, Poland
| | - Anna Śmiech
- Department of Pathological Anatomy, Faculty of Veterinary Medicine, University of Life Sciences, 20-033 Lublin, Poland
| |
Collapse
|
22
|
Ling K, Men F, Wang WC, Zhou YQ, Zhang HW, Ye DW. Carbon Monoxide and Its Controlled Release: Therapeutic Application, Detection, and Development of Carbon Monoxide Releasing Molecules (CORMs). J Med Chem 2017; 61:2611-2635. [PMID: 28876065 DOI: 10.1021/acs.jmedchem.6b01153] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Carbon monoxide (CO) is attracting increasing attention because of its role as a gasotransmitter with cytoprotective and homeostatic properties. Carbon monoxide releasing molecules (CORMs) are spatially and temporally controlled CO releasers that exhibit superior and more effective pharmaceutical traits than gaseous CO because of their chemistry and structure. Experimental and preclinical research in animal models has shown the therapeutic potential of inhaled CO and CORMs, and the biological effects of CO and CORMs have also been observed in preclinical trials via the genetic modulation of heme oxygenase-1 (HO-1). In this review, we describe the pharmaceutical use of CO and CORMs, methods of detecting CO release, and developments in CORM design and synthesis. Many valuable clinical CORMs formulated using macromolecules and nanomaterials are also described.
Collapse
Affiliation(s)
- Ken Ling
- Cancer Center, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China.,Department of Anesthesiology, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China
| | - Fang Men
- College of Chemistry and Molecular Sciences , Wuhan University , Wuhan 430072 , China
| | - Wei-Ci Wang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China
| | - Ya-Qun Zhou
- Anesthesiology Institute, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China
| | - Hao-Wen Zhang
- Cancer Center, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China
| | - Da-Wei Ye
- Cancer Center, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China
| |
Collapse
|
23
|
Chifman J, Arat S, Deng Z, Lemler E, Pino JC, Harris LA, Kochen MA, Lopez CF, Akman SA, Torti FM, Torti SV, Laubenbacher R. Activated Oncogenic Pathway Modifies Iron Network in Breast Epithelial Cells: A Dynamic Modeling Perspective. PLoS Comput Biol 2017; 13:e1005352. [PMID: 28166223 PMCID: PMC5293201 DOI: 10.1371/journal.pcbi.1005352] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 01/08/2017] [Indexed: 12/21/2022] Open
Abstract
Dysregulation of iron metabolism in cancer is well documented and it has been suggested that there is interdependence between excess iron and increased cancer incidence and progression. In an effort to better understand the linkages between iron metabolism and breast cancer, a predictive mathematical model of an expanded iron homeostasis pathway was constructed that includes species involved in iron utilization, oxidative stress response and oncogenic pathways. The model leads to three predictions. The first is that overexpression of iron regulatory protein 2 (IRP2) recapitulates many aspects of the alterations in free iron and iron-related proteins in cancer cells without affecting the oxidative stress response or the oncogenic pathways included in the model. This prediction was validated by experimentation. The second prediction is that iron-related proteins are dramatically affected by mitochondrial ferritin overexpression. This prediction was validated by results in the pertinent literature not used for model construction. The third prediction is that oncogenic Ras pathways contribute to altered iron homeostasis in cancer cells. This prediction was validated by a combination of simulation experiments of Ras overexpression and catalase knockout in conjunction with the literature. The model successfully captures key aspects of iron metabolism in breast cancer cells and provides a framework upon which more detailed models can be built. Iron is required for cellular metabolism and growth, but can be toxic due to its ability to cause high oxidative stress and consequently DNA damage. To prevent damage, all organisms that require iron have developed mechanisms to tightly control iron levels. Dysregulation of iron metabolism is detrimental and can contribute to a wide range of diseases, including cancer. This paper presents a predictive mathematical model of iron regulation linked to iron utilization, oxidative stress, and the oncogenic response specific to normal breast epithelial cells. The model uses a discrete modeling framework to generate novel biological hypotheses for an investigation of how normal breast cells become malignant cells, capturing a breast cancer phenotype of iron homeostasis through overexpression and knockout simulations. The new biology discovered is (1) IRP2 overexpression alters the iron homeostasis pathway in breast cells, without affecting the oxidative stress response or oncogenic pathways, (2) an activated oncogenic pathway disrupts iron regulation in breast cancer cells.
Collapse
Affiliation(s)
- Julia Chifman
- Department of Mathematics and Statistics, American University, Washington, DC, USA
| | - Seda Arat
- The Jackson Laboratory, Bar Harbor, ME, USA
| | - Zhiyong Deng
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, USA
| | - Erica Lemler
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, USA
| | - James C. Pino
- Chemical and Physical Biology Graduate Program, Vanderbilt University, Nashville, TN, USA
| | - Leonard A. Harris
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Michael A. Kochen
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | - Carlos F. Lopez
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
- Center for Quantitative Science, Vanderbilt University, Nashville, TN, USA
| | - Steven A. Akman
- Cancer Program, Roper St Francis HealthCare, Charleston, SC, USA
| | - Frank M. Torti
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Suzy V. Torti
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, USA
| | - Reinhard Laubenbacher
- Center for Quantitative Medicine, University of Connecticut Health Center, Farmington, CT, USA
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- * E-mail:
| |
Collapse
|
24
|
Role of PTEN in Oxidative Stress and DNA Damage in the Liver of Whole-Body Pten Haplodeficient Mice. PLoS One 2016; 11:e0166956. [PMID: 27893783 PMCID: PMC5125655 DOI: 10.1371/journal.pone.0166956] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 11/07/2016] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes (T2DM) and obesity are frequently associated with non-alcoholic fatty liver disease (NAFLD) and with an elevated cancer incidence. The molecular mechanisms of carcinogenesis in this context are only partially understood. High blood insulin levels are typical in early T2DM and excessive insulin can cause elevated reactive oxygen species (ROS) production and genomic instability. ROS are important for various cellular functions in signaling and host defense. However, elevated ROS formation is thought to be involved in cancer induction. In the molecular events from insulin receptor binding to genomic damage, some signaling steps have been identified, pointing at the PI3K/AKT pathway. For further elucidation Phosphatase and Tensin homolog (Pten), a tumour suppressor phosphatase that plays a role in insulin signaling by negative regulation of PI3K/AKT and its downstream targets, was investigated here. Dihydroethidium (DHE) staining was used to detect ROS formation in immortalized human hepatocytes. Comet assay and micronucleus test were performed to investigate genomic damage in vitro. In liver samples, DHE staining and western blot detection of HSP70 and HO-1 were performed to evaluate oxidative stress response. DNA double strand breaks (DSBs) were detected by immunohistostaining. Inhibition of PTEN with the pharmacologic inhibitor VO-OHpic resulted in increased ROS production and genomic damage in a liver cell line. Knockdown of Pten in a mouse model yielded increased oxidative stress levels, detected by ROS levels and expression of the two stress-proteins HSP70 and HO-1 and elevated genomic damage in the liver, which was significant in mice fed with a high fat diet. We conclude that PTEN is involved in oxidative stress and genomic damage induction in vitro and that this may also explain the in vivo observations. This further supports the hypothesis that the PI3K/AKT pathway is responsible for damaging effects of high levels of insulin.
Collapse
|
25
|
Enhancement of DEN-induced liver tumorigenesis in heme oxygenase-1 G143H mutant transgenic mice. Biochem Biophys Res Commun 2016; 481:169-175. [PMID: 27810363 DOI: 10.1016/j.bbrc.2016.10.148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 10/30/2016] [Indexed: 12/25/2022]
Abstract
Heme oxygenase (HO) is the rate-limiting enzyme in heme metabolism. HO-1 exhibits anti-oxidative and anti-inflammatory function via the actions of its metabolite, respectively. A growing body of evidence demonstrates that HO-1 is implicated in the pathogenesis and progression of several types of cancer. However, whether HO-1 takes part in healthy-premalignant-malignant transformation is still undefined. In this study, we took advantage of transgenic mice which over-expressed HO-1 dominant negative mutant (HO-1 G143H) and observed its susceptibility to DEN-induced hepatocarcinogenesis. Our results indicate that HO-1 G143H mutant accelerates the progression of tumorigenesis and tumor growth. The mechanism is closely related to enhancement of ROS production which induce more hepatocytes death and secretion of inflammatory cytokines, proliferation of surviving hepatocytes. Our result provides the direct evidence that HO-1 plays an important protective role in liver carcinogenesis. Alternatively, we suggest the possible explanation on effect of HO-1 promoter polymorphism which involved in tumorigenesis.
Collapse
|
26
|
Zou C, Zou C, Cheng W, Li Q, Han Z, Wang X, Jin J, Zou J, Liu Z, Zhou Z, Zhao W, Du Z. Heme oxygenase-1 retards hepatocellular carcinoma progression through the microRNA pathway. Oncol Rep 2016; 36:2715-2722. [PMID: 27571925 DOI: 10.3892/or.2016.5056] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 07/25/2016] [Indexed: 11/06/2022] Open
Abstract
Heme metabolism system is involved in microRNA (miRNA) biogenesis. The complicated interplay between heme oxygenase-1 (HO-1) and miRNA has been observed in various tissues and diseases, including human malignancy. In the present study, our data showed that stable HO-1 overexpression in hepatocellular carcinoma (HCC) cells downregulated several oncomiRs. The most stably downregulated are miR-30d and miR-107. Iron, one of HO-1 catalytic products, was an important mediator in this regulation. Cell function analysis demonstrated that HO-1 inhibited the proliferation and metastasis of HepG2 cells, whereas miR-30d/miR-107 improved the proliferative and migratory ability of HepG2 cells. The beneficial effect of HO-1 in HCC inhibition could be reversed by upregulating miR-30d and miR-107. Akt and ERK pathways may be involved in the regulation of HO-1/miR-30d/miR-107 in HCC. These data indicate that HO-1 significantly suppresses HCC progression by regulating the miR-30d/miR-107 level, suggesting miR-30d/miR-107 regulation as a new molecular mechanism of HO-1 anticancer effect.
Collapse
Affiliation(s)
- Chaoxia Zou
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Chendan Zou
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Wanpeng Cheng
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Qiang Li
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Zhongjing Han
- Department of Hemopathology, General Hospital of Daqing Oil Field, Daqing, Heilongjiang, P.R. China
| | - Xiaona Wang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Jianfeng Jin
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Jiaqi Zou
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Zhiyan Liu
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Zhongqiu Zhou
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Weiming Zhao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, P.R. China
| | - Zhimin Du
- Institute of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, P.R. China
| |
Collapse
|
27
|
Heme oxygenase-1 induction attenuates imiquimod-induced psoriasiform inflammation by negative regulation of Stat3 signaling. Sci Rep 2016; 6:21132. [PMID: 26893174 PMCID: PMC4759695 DOI: 10.1038/srep21132] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/19/2016] [Indexed: 12/24/2022] Open
Abstract
Heme oxygenase-1 (HO-1), a stress-inducible protein with a potential anti-inflammatory effect, plays an important role in skin injury and wound healing. However, the function of HO-1 in cutaneous inflammatory diseases, such as psoriasis, remains unknown. The abnormal activation of Stat3, a known transcription factor that induces inflammation and regulates cell differentiation, is directly involved in the pathogenesis and development of psoriasis. Hence, targeting Stat3 is potentially beneficial in the treatment of psoriasis. In this study, HO-1 activation significantly alleviated the disease-related pathogenesis abnormality. To determine the mechanism by which HO-1 exerts immune protection on Th17-related cytokines, IL6/IL22-induced Stat3 activation was significantly suppressed, accompanied by decreased cell proliferation and reversed abnormal cell proliferation. Importantly, HO-1-induced Stat3 suppression was mediated through the activation of protein tyrosine phosphatase SHP-1. Overall, our study provides direct evidence indicating that HO-1 might be a useful therapeutic target for psoriasis. SHP-1-mediated suppression of Stat3 activation after HO-1 activation is a unique molecular mechanism for the regulation of Stat3 activation.
Collapse
|
28
|
Abstract
The three endogenous gaseous transmitters - nitric oxide (NO), carbon monoxide (CO) and hydrogen sulfide (H2S) - regulate a number of key biological functions. Emerging data have revealed several new mechanisms for each of these three gasotransmitters in tumour biology. It is now appreciated that they show bimodal pharmacological character in cancer, in that not only the inhibition of their biosynthesis but also elevation of their concentration beyond a certain threshold can exert anticancer effects. This Review discusses the role of each gasotransmitter in cancer and the effects of pharmacological agents - some of which are in early-stage clinical studies - that modulate the levels of each gasotransmitter. A clearer understanding of the pharmacological character of these three gases and the mechanisms underlying their biological effects is expected to guide further clinical translation.
Collapse
|
29
|
Chen H, Wang X, Zhao Q, Zhang Z, Ye X, Hua F, Cui G. Dual effects of heme oxygenase-1 on astrocyte injury induced by heminin vitro. Brain Inj 2015; 30:36-42. [DOI: 10.3109/02699052.2015.1084648] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
30
|
Abstract
BACKGROUND The KEAP1-Nrf2 antioxidant signaling pathway is important in protecting liver from various insults. However, little is known about the expression of Nrf2-related genes in human liver in different diseases. METHODS This study utilized normal donor liver tissues (n=35), samples from patients with hepatocellular carcinoma (HCC, n=24), HBV-related cirrhosis (n=27), alcoholic cirrhosis (n=5) and end-stage liver disease (n=13). All of the liver tissues were from the Oriental Liver Transplant Center, Beijing, China. The expressions of Nrf2 and Nrf2-related genes, including its negative regulator Kelch-like ECH-associated protein 1 (KEAP1), its targeted gene NAD(P)H-quinone oxidoreductase 1 (NQO1), glutamate-cysteine ligase catalytic subunit (GCLC) and modified subunit (GCLM), heme oxygenase 1 (HO-1) and peroxiredoxin-1 (PRDX1) were evaluated. RESULTS The expression of Nrf2 was decreased in HCC, increased in alcoholic cirrhosis and end-stage liver disease. The expression of KEAP1 was increased in all of the liver samples. The most notable finding was the increased expression of NQO1 in HCC (18-fold), alcoholic cirrhosis (6-fold), end-stage liver disease (5-fold) and HBV-related cirrhosis (3-fold). Peri-HCC also had 4-fold higher NQO1 mRNA as compared to the normal livers. GCLC mRNA levels were lower only in HCC, as compared to the normal livers and peri-HCC tissues. GCLM mRNA levels were higher in HBV-related cirrhosis and end-stage liver disease. HO-1 mRNA levels were increased in all liver tissues except for HCC. Peri-HCC had higher PRDX1 mRNA levels compared with HCC and normal livers. CONCLUSION Nrf2 and Nrf2-related genes are aberrantly expressed in the liver in different diseases and the increase of NQO1 was the most notable finding, especially in HCC.
Collapse
|
31
|
Tan JY, Chen JL, Huang X, Yuan CL. Screening and verification of proteins that interact with HSPC238. Oncol Rep 2015; 34:3097-103. [PMID: 26398855 DOI: 10.3892/or.2015.4289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 07/14/2015] [Indexed: 11/06/2022] Open
Abstract
HSPC238 is a recently identified tumor suppressor and demonstrates ubiquitin ligase E3 enzyme activity. HSPC238 was found to be significantly downregulated in human hepatocellular carcinoma (HCC) in vivo and to inhibit the proliferation and invasion of hepatoma cells in vitro; however, the underlying molecular mechanism is largely unknown. In the present study, we screened for and identified proteins that physically interact with HSPC238. A bait vector for yeast two-hybrid was constructed with human HSPC238 gene cDNA. Yeast two-hybrid screening was performed using a human fetal liver cDNA library. Multiple reporter gene assays, DNA sequencing and BLAST comparison analysis were performed on positive clones. Protein interaction of screened candidates with HSPC238 was further validated by confocal microscopy, co-immunoprecipitation and pull-down assays. Yeast two-hybrid screening demonstrated 124 positive clones. Multiple reporter gene assays with LacZ, HIS and ADE2 selective media identified 12 genes. Further co-localization, co-immunoprecipitation and pull-down assays demonstrated that HMOX1, RPS27A, ubiquitinB and MT2A interacted with HSPC238. These four proteins are involved in tumor development and progression, and are associated with the ubiquitin-proteasome pathway. Our results suggest that HSPC238 may play a tumor suppressor role and interact with these proteins via the ubiquitin-proteasome pathway. The identification and validation of proteins interacting with HSP238 may lead to the discovery of novel mechanisms through which HSPC238 suppresses tumorigenesis in human hepatocellular carcinoma.
Collapse
Affiliation(s)
- Jia-Yu Tan
- Central Intensive Care Unit, Bo'ai Hospital of Zhongshan City Affiliated with Southern Medical University, Zhongshan 528403, P.R. China
| | - Jing-Lin Chen
- Prenatal Diagnosis Center, Bo'ai Hospital of Zhongshan City Affiliated with Southern Medical University, Zhongshan 528403, P.R. China
| | - Xiang Huang
- Prenatal Diagnosis Center, Bo'ai Hospital of Zhongshan City Affiliated with Southern Medical University, Zhongshan 528403, P.R. China
| | - Chun-Lei Yuan
- Department of Laboratory Medicine, Bo'ai Hospital of Zhongshan City Affiliated with Southern Medical University, Zhongshan 528403, P.R. China
| |
Collapse
|
32
|
Lee HN, Jin HO, Park JA, Kim JH, Kim JY, Kim B, Kim W, Hong SE, Lee YH, Chang YH, Hong SI, Hong YJ, Park IC, Surh YJ, Lee JK. Heme oxygenase-1 determines the differential response of breast cancer and normal cells to piperlongumine. Mol Cells 2015; 38:327-35. [PMID: 25813625 PMCID: PMC4400307 DOI: 10.14348/molcells.2015.2235] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/09/2014] [Accepted: 12/22/2014] [Indexed: 12/30/2022] Open
Abstract
Piperlongumine, a natural alkaloid isolated from the long pepper, selectively increases reactive oxygen species production and apoptotic cell death in cancer cells but not in normal cells. However, the molecular mechanism underlying piperlongumine-induced selective killing of cancer cells remains unclear. In the present study, we observed that human breast cancer MCF-7 cells are sensitive to piperlongumine-induced apoptosis relative to human MCF-10A breast epithelial cells. Interestingly, this opposing effect of piperlongumine appears to be mediated by heme oxygenase-1 (HO-1). Piperlongumine upregulated HO-1 expression through the activation of nuclear factor-erythroid-2-related factor-2 (Nrf2) signaling in both MCF-7 and MCF-10A cells. However, knockdown of HO-1 expression and pharmacological inhibition of its activity abolished the ability of piperlongumine to induce apoptosis in MCF-7 cells, whereas those promoted apoptosis in MCF-10A cells, indicating that HO-1 has anti-tumor functions in cancer cells but cytoprotective functions in normal cells. Moreover, it was found that piperlongumine-induced Nrf2 activation, HO-1 expression and cancer cell apoptosis are not dependent on the generation of reactive oxygen species. Instead, piperlongumine, which bears electrophilic α,β-unsaturated carbonyl groups, appears to inactivate Kelch-like ECH-associated protein-1 (Keap1) through thiol modification, thereby activating the Nrf2/HO-1 pathway and subsequently upregulating HO-1 expression, which accounts for piperlongumine-induced apoptosis in cancer cells. Taken together, these findings suggest that direct interaction of piperlongumine with Keap1 leads to the upregulation of Nrf2-mediated HO-1 expression, and HO-1 determines the differential response of breast normal cells and cancer cells to piperlongumine.
Collapse
Affiliation(s)
- Ha-Na Lee
- KIRAMS Radiation Biobank, Korea Institute of Radiological and Medical Sciences, Seoul 139-709,
Korea
| | - Hyeon-Ok Jin
- KIRAMS Radiation Biobank, Korea Institute of Radiological and Medical Sciences, Seoul 139-709,
Korea
| | - Jin-Ah Park
- KIRAMS Radiation Biobank, Korea Institute of Radiological and Medical Sciences, Seoul 139-709,
Korea
| | - Jin-Hee Kim
- KIRAMS Radiation Biobank, Korea Institute of Radiological and Medical Sciences, Seoul 139-709,
Korea
| | - Ji-Young Kim
- KIRAMS Radiation Biobank, Korea Institute of Radiological and Medical Sciences, Seoul 139-709,
Korea
| | - BoRa Kim
- KIRAMS Radiation Biobank, Korea Institute of Radiological and Medical Sciences, Seoul 139-709,
Korea
| | - Wonki Kim
- College of Pharmacy, Seoul National University, Seoul 151-742,
Korea
| | - Sung-Eun Hong
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-709,
Korea
| | - Yun-Han Lee
- Department of Radiation Oncology, College of Medicine, Yonsei University, Seoul 120-752,
Korea
| | - Yoon Hwan Chang
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 139-709,
Korea
| | - Seok-Il Hong
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 139-709,
Korea
| | - Young Jun Hong
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 139-709,
Korea
| | - In-Chul Park
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-709,
Korea
| | - Young-Joon Surh
- College of Pharmacy, Seoul National University, Seoul 151-742,
Korea
| | - Jin Kyung Lee
- KIRAMS Radiation Biobank, Korea Institute of Radiological and Medical Sciences, Seoul 139-709,
Korea
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 139-709,
Korea
| |
Collapse
|
33
|
Yang N, Jiang Y, Zhang H, Sun B, Hou C, Zheng J, Liu Y, Zuo P. Active targeting docetaxel-PLA nanoparticles eradicate circulating lung cancer stem-like cells and inhibit liver metastasis. Mol Pharm 2014; 12:232-9. [PMID: 25418453 DOI: 10.1021/mp500568z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Lung cancer is the major cause of cancer related lethality worldwide, and metastasis to distant organs is the pivotal cause of death for the vast majority of lung cancer patients. Accumulated evidence indicates that lung cancer stem-like cells (CSLCs) play important roles in metastagenesis, and these circulating CSLCs may be important targets to inhibit the subsequent metastasis. The present study was aimed at establishing CSLC-targeting polylactic acid (PLA) encapsulated docetaxel nanoparticles for antimetastatic therapy. Cyclic binding peptides were screened on CSLCs in vitro and the peptide CVKTPAQSC exhibiting high specific binding ability to pulmonary adenocarcinoma tissue was subsequently conjugated to the nanoparticles loaded with docetaxel (NDTX). Antimetastatic effect of CSLC-targeting nanoparticles loaded with docetaxel (TNDTX) was evaluated in a nude mouse model of liver metastasis. Results showed that, in the absence of targeting peptide, NDTX hardly exhibited any antimetastatic effect. However, TNDTX treatment significantly decreased the metastatic tumor area in the nude mouse liver. Histopathological and serological results also confirmed the antimetastatic efficacy of TNDTX. To our knowledge, this is the first report on establishing a CSLC-based strategy for lung cancer metastatic treatment, and we hope this will offer a potential therapeutic approach for management of metastatic lung cancer.
Collapse
Affiliation(s)
- Nan Yang
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College , 100005, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Myeloperoxidase oxidized LDL interferes with endothelial cell motility through miR-22 and heme oxygenase 1 induction: possible involvement in reendothelialization of vascular injuries. Mediators Inflamm 2014; 2014:134635. [PMID: 25530680 PMCID: PMC4233670 DOI: 10.1155/2014/134635] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 09/09/2014] [Indexed: 01/19/2023] Open
Abstract
Cardiovascular disease linked to atherosclerosis is the leading cause of death worldwide. Atherosclerosis is mainly linked to dysfunction in vascular endothelial cells and subendothelial accumulation of oxidized forms of LDL. In the present study, we investigated the role of myeloperoxidase oxidized LDL (Mox-LDL) in endothelial cell dysfunction. We studied the effect of proinflammatory Mox-LDL treatment on endothelial cell motility, a parameter essential for normal vascular processes such as angiogenesis and blood vessel repair. This is particularly important in the context of an atheroma plaque, where vascular wall integrity is affected and interference with its repair could contribute to progression of the disease. We investigated in vitro the effect of Mox-LDL on endothelial cells angiogenic properties and we also studied the signalling pathways that could be affected by analysing Mox-LDL effect on the expression of angiogenesis-related genes. We report that Mox-LDL inhibits endothelial cell motility and tubulogenesis through an increase in miR-22 and heme oxygenase 1 expression. Our in vitro data indicate that Mox-LDL interferes with parameters associated with angiogenesis. They suggest that high LDL levels in patients would impair their endothelial cell capacity to cope with a damaged endothelium contributing negatively to the progression of the atheroma plaque.
Collapse
|
35
|
Oldani G, Crowe LA, Orci LA, Slits F, Rubbia-Brandt L, de Vito C, Morel P, Mentha G, Berney T, Vallée JP, Lacotte S, Toso C. Pre-retrieval reperfusion decreases cancer recurrence after rat ischemic liver graft transplantation. J Hepatol 2014; 61:278-85. [PMID: 24713189 DOI: 10.1016/j.jhep.2014.03.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 02/28/2014] [Accepted: 03/27/2014] [Indexed: 01/14/2023]
Abstract
BACKGROUND & AIMS Liver transplantation from marginal donors is associated with ischemia/reperfusion (I/R) lesions, which may increase the risk of post-transplant hepatocellular carcinoma (HCC) recurrence. Graft reperfusion prior to retrieval (as for extracorporeal membrane oxygenation--ECMO) can prevent I/R lesions. The impact of I/R on the risk of cancer recurrence was assessed on a syngeneic Fischer-rat liver transplantation model. METHODS HCC cells were injected into the vena porta of all recipients at the end of an orthotopic liver transplantation (OLT). Control donors were standard heart-beating, ischemic ones (ISC), underwent 10 min or 30 min inflow liver clamping prior to retrieval, and ischemic/reperfused (ISC/R) donors underwent 2h liver reperfusion after the clamping. RESULTS I/R lesions were confirmed in the ISC group, with the presence of endothelial and hepatocyte injury, and increased liver function tests. These lesions were in part reversed by the 2h reperfusion in the ISC/R group. HCC growth was higher in the 10 min and 30 min ISC recipients (p = 0.018 and 0.004 vs. control, as assessed by MRI difference between weeks one and two), and was prevented in the ISC/Rs (p = 0.04 and 0.01 vs. ISC). These observations were associated with a stronger pro-inflammatory cytokine profile in the ISC recipients only, and the expression of hypoxia and HCC growth-enhancer genes, including Hmox1, Hif1a and Serpine1. CONCLUSIONS This experiment suggests that ischemia/reperfusion lesions lead to an increased risk of post-transplant HCC recurrence and growth. This observation can be reversed by graft reperfusion prior to retrieval.
Collapse
Affiliation(s)
- Graziano Oldani
- Divisions of Transplant and Abdominal Surgery, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Surgery, University of Pavia, Italy
| | - Lindsey A Crowe
- Division of Radiology, Department of Medical Imaging, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Lorenzo A Orci
- Divisions of Transplant and Abdominal Surgery, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Florence Slits
- Divisions of Transplant and Abdominal Surgery, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Laura Rubbia-Brandt
- Division of Clinical Pathology, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Claudio de Vito
- Division of Clinical Pathology, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Philippe Morel
- Divisions of Transplant and Abdominal Surgery, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Gilles Mentha
- Divisions of Transplant and Abdominal Surgery, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Thierry Berney
- Divisions of Transplant and Abdominal Surgery, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jean-Paul Vallée
- Division of Radiology, Department of Medical Imaging, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Stéphanie Lacotte
- Divisions of Transplant and Abdominal Surgery, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Christian Toso
- Divisions of Transplant and Abdominal Surgery, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
36
|
Antiproliferative effects of carbon monoxide on pancreatic cancer. Dig Liver Dis 2014; 46:369-75. [PMID: 24433995 DOI: 10.1016/j.dld.2013.12.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 11/26/2013] [Accepted: 12/04/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND Carbon monoxide, the gaseous product of heme oxygenase, is a signalling molecule with a broad spectrum of biological activities. The aim of this study was to investigate the effects of carbon monoxide on proliferation of human pancreatic cancer. METHODS In vitro studies were performed on human pancreatic cancer cells (CAPAN-2, BxPc3, and PaTu-8902) treated with a carbon monoxide-releasing molecule or its inactive counterpart, or exposed to carbon monoxide gas (500 ppm/24h). For in vivo studies, pancreatic cancer cells (CAPAN-2/PaTu-8902) were xenotransplanted subcutaneously into athymic mice, subsequently treated with carbon monoxide-releasing molecule (35 mg/kg b.w. i.p./day), or exposed to safe doses of carbon monoxide (500 ppm 1h/day; n = 6 in each group). RESULTS Both carbon monoxide-releasing molecule and carbon monoxide exposure significantly inhibited proliferation of human pancreatic cancer cells (p<0.05). A substantial decrease in Akt phosphorylation was observed in carbon monoxide-releasing molecule compared with inactive carbon monoxide-releasing molecule treated cancer cells (by 30-50%, p<0.05). Simultaneously, carbon monoxide-releasing molecule and carbon monoxide exposure inhibited tumour proliferation and microvascular density of xenotransplanted tumours (p<0.01), and doubled the survival rates (p<0.005). Exposure of mice to carbon monoxide led to an almost 3-fold increase in carbon monoxide content in tumour tissues (p=0.006). CONCLUSION These data suggest a new biological function for carbon monoxide in carcinogenesis, and point to the potential chemotherapeutic/chemoadjuvant use of carbon monoxide in pancreatic cancer.
Collapse
|
37
|
Yang BC, Yang ZH, Pan XJ, Wang LM, Liu XY, Zhu MX, Xie JP. Transcript profiling analysis of in vitro cultured THP-1 cells after exposure to crotonaldehyde. J Toxicol Sci 2014; 39:487-97. [DOI: 10.2131/jts.39.487] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Bi-cheng Yang
- Department of Radiation Toxicology and Oncology, Beijing Institute of Radiation Medicine
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences
| | - Zhi-hua Yang
- Department of Radiation Toxicology and Oncology, Beijing Institute of Radiation Medicine
| | - Xiu-jie Pan
- Department of Radiation Toxicology and Oncology, Beijing Institute of Radiation Medicine
| | - Li-meng Wang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences
| | - Xing-yu Liu
- Beijing Work Station, Technology Center of Shanghai Tobacco Corporation
| | - Mao-xiang Zhu
- Department of Radiation Toxicology and Oncology, Beijing Institute of Radiation Medicine
| | | |
Collapse
|
38
|
The induction of heme oxygenase-1 suppresses heat shock protein 90 and the proliferation of human breast cancer cells through its byproduct carbon monoxide. Toxicol Appl Pharmacol 2013; 274:55-62. [PMID: 24211270 DOI: 10.1016/j.taap.2013.10.027] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 10/09/2013] [Accepted: 10/28/2013] [Indexed: 11/24/2022]
Abstract
Heme oxygenase (HO)-1 is an oxidative stress-response enzyme which catalyzes the degradation of heme into bilirubin, ferric ion, and carbon monoxide (CO). Induction of HO-1 was reported to have antitumor activity; the inhibitory mechanism, however, is still unclear. In the present study, we found that treatment with [Ru(CO)3Cl2]2 (RuCO), a CO-releasing compound, reduced the growth of human MCF7 and MDA-MB-231 breast cancer cells. Analysis of growth-related proteins showed that treatment with RuCO down-regulated cyclinD1, CDK4, and hTERT protein expressions. Interestingly, RuCO treatment resulted in opposite effects on wild-type and mutant p53 proteins. These results were similar to those of cells treated with geldanamycin (a heat shock protein (HSP)90 inhibitor), suggesting that RuCO might affect HSP90 activity. Moreover, RuCO induced mutant p53 protein destabilization accompanied by promotion of ubiquitination and proteasome degradation. The induction of HO-1 by cobalt protoporphyrin IX (CoPP) showed consistent results, while the addition of tin protoporphyrin IX (SnPP), an HO-1 enzymatic inhibitor, diminished the RuCO-mediated effect. RuCO induction of HO-1 expression was reduced by a p38 mitogen-activated protein kinase inhibitor (SB203580). Additionally, treatment with a chemopreventive compound, curcumin, induced HO-1 expression accompanied with reduction of HSP90 client protein expression. The induction of HO-1 by curcumin inhibited 12-O-tetradecanoyl-13-acetate (TPA)-elicited matrix metalloproteinase-9 expression and tumor invasion. In conclusion, we provide novel evidence underlying HO-1's antitumor mechanism. CO, a byproduct of HO-1, suppresses HSP90 protein activity, and the induction of HO-1 may possess potential as a cancer therapeutic.
Collapse
|
39
|
Othman MS, Aref AM, Mohamed AA, Ibrahim WA. Serum Levels of Interleukin-6 and Interleukin-10 as Biomarkers for Hepatocellular Carcinoma in Egyptian Patients. ISRN HEPATOLOGY 2013; 2013:412317. [PMID: 27335826 PMCID: PMC4890868 DOI: 10.1155/2013/412317] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 08/12/2013] [Indexed: 02/07/2023]
Abstract
Interleukin-10 (IL-10) and interleukin-6 (IL-6) have been reported to be related to hepatocellular carcinoma (HCC) prognosis. This study aimed to investigate the clinical usefulness of serum levels of IL-6 and IL-10 as biomarkers for HCC among high-risk patients. Materials and Methods. 80 individuals were enrolled in this study; they were categorized into 4 groups: group 1 healthy individuals (NC) (n = 20), group 2 chronic hepatitis C virus (HCV) patients (n = 20), group 3 cirrhotic patients (LC) (n = 20), and HCC group (n = 20). Using ELISA technique serum levels of IL-6, IL-10, and alpha fetoprotein (AFP) were evaluated in all groups. Results. The mean serum levels of IL-6 were significantly higher in HCC than in LC, HCV, and NC groups (13.99 ± 1.80, 7.49 ± 0.43, 5.78 ± 0.74, and 2.57 ± 0.31), respectively (P < 0.05); also the serum levels of IL-10 were significantly higher in HCC compared with LC, HCV, and NC groups (13.69 ± 1.89, 7.37 ± 0.53, 5.18 ± 0.6, and 3.31 ± 0.42) (P < 0.05). We also found that the tumor size is correlated strongly with IL-6 and IL-10 levels (r = 0.925, P < 0.001; r = 0.821, P < 0.001), respectively. Conclusion. The combination of those markers may help to identify a group of HCC patients with low AFP.
Collapse
Affiliation(s)
- Mohamed S. Othman
- Biochemistry Department, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Giza 11787, Egypt
| | - Ahmed M. Aref
- Biology Department, Faculty of Dentistry, October University for Modern Sciences and Arts (MSA), Giza 11787, Egypt
| | - Amal A. Mohamed
- Biochemsitry Department, National Hepatology and Tropical Medicine Research Institute, Fom El-Khalig, Cairo 11796, Egypt
| | | |
Collapse
|
40
|
Luo J, Zhou X, Ge X, Liu P, Cao J, Lu X, Ling Y, Zhang S. Upregulation of Ying Yang 1 (YY1) suppresses esophageal squamous cell carcinoma development through heme oxygenase-1. Cancer Sci 2013; 104:1544-51. [PMID: 23919806 DOI: 10.1111/cas.12248] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 07/16/2013] [Accepted: 07/26/2013] [Indexed: 12/24/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the deadliest malignancies worldwide. Ying Yang 1 (YY1), a ubiquitously expressed GLI-Krüppel zinc finger transcription factor, plays a regulatory role in a variety of fundamental biological processes, such as embryonic development, growth, apoptosis, differentiation and oncogenic transformation. The purpose of this study was to investigate the expression of YY1 in normal and cancerous esophageal tissues and its function in ESCC development. We found that the expression of YY1 mRNA was significantly increased in the tumor tissues, compared with the para-tissues or normal esophageal tissues. The increased expression of YY1 in tumor samples was further confirmed by immunohistochemistry. Furthermore, the overexpression of YY1 conferred radioresistance to the ESCC TE-1 cells and resulted in markedly reduced cell proliferation. Accordingly, the small interfering RNA-mediated silencing of YY1 expression in TE-1 cells resulted in increased proliferation by enhancing the binding of P21 to Cyclin D1 and CDK4, a protein complex known to mediate cell cycle progression. Moreover, besides P21, heme oxygenase-1 (HO-1) was identified as a YY1 downstream effector, as YY1 stimulated HO-1 expression in esophageal cancer cells. YY1 mediated biological function through transcription of HO-1. Forced expression of HO-1 could moderately suppress proliferation of TE-1 cells. The expression of YY1 significantly correlated with that of HO-1 in ESCC tissues. Taken together, we demonstrated overexpression of YY1 in esophageal carcinoma and identified HO-1 as its target.
Collapse
Affiliation(s)
- Judong Luo
- Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou, China; Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Hu JL, Xiao L, Li ZY, Wang Q, Chang Y, Jin Y. Upregulation of HO-1 is accompanied by activation of p38MAPK and mTOR in human oesophageal squamous carcinoma cells. Cell Biol Int 2013; 37:584-92. [PMID: 23412940 DOI: 10.1002/cbin.10075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 02/06/2013] [Indexed: 12/31/2022]
Affiliation(s)
- Jian-Li Hu
- Cancer Centre, Union Hospital, Huazhong University of Science and Technology; 1277 Jiefangdadao Jianghan District, Wuhan; Hubei; 430022; PR China
| | - Lan Xiao
- Department of Obstetrics and Gynecology; First Affiliated Hospital, An Hui Medical College; 218 Jixi Road, Hefei; AnHui; 230022; PR China
| | - Zhen-Yun Li
- Cancer Centre, Union Hospital, Huazhong University of Science and Technology; 1277 Jiefangdadao Jianghan District, Wuhan; Hubei; 430022; PR China
| | - Qiong Wang
- Cancer Centre, Union Hospital, Huazhong University of Science and Technology; 1277 Jiefangdadao Jianghan District, Wuhan; Hubei; 430022; PR China
| | - Yu Chang
- Cancer Centre, Union Hospital, Huazhong University of Science and Technology; 1277 Jiefangdadao Jianghan District, Wuhan; Hubei; 430022; PR China
| | - Yi Jin
- Laboratory Department; Union Hospital, Huazhong University of Science and Technology; 1277 Jiefangdadao Jianghan District, Wuhan; Hubei; 430022; PR China
| |
Collapse
|
42
|
MicroRNA-25 functions as a potential tumor suppressor in colon cancer by targeting Smad7. Cancer Lett 2013; 335:168-74. [PMID: 23435373 DOI: 10.1016/j.canlet.2013.02.029] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 02/03/2013] [Accepted: 02/05/2013] [Indexed: 12/22/2022]
Abstract
Because it is a member of the miR-106b~25 cluster, microRNA-25 (miR-25) is known to be dysregulated in human cancers. However, the expression and role of miR-25 in colon cancer remain unclear. In this study, miR-25 was found to be down-regulated in human colon cancer tissues when compared to those in matched, non-neoplastic mucosa tissues. Functional studies revealed that restoration of miR-25 expression inhibited cell proliferation and migration. In contrast, miR-25 inhibition could promote the proliferation and migratory ability of cells. Stable over-expression of miR-25 also suppressed the growth of colon cancer-cell xenografts in vivo. Furthermore, bioinformatic predictions and experimental validation were used to identify Smad7 as a direct target of miR-25. Functional reverse experiments indicated that the antitumor effects of miR-25 were probably mediated by its repression of Smad7. These results suggest that miR-25 may function as a tumor suppressor by targeting Smad7 in colon cancer. Thus, miR-25 may serve as a potential therapeutic agent or target for cancer therapy.
Collapse
|
43
|
Chen HW, Chao CY, Lin LL, Lu CY, Liu KL, Lii CK, Li CC. Inhibition of matrix metalloproteinase-9 expression by docosahexaenoic acid mediated by heme oxygenase 1 in 12-O-tetradecanoylphorbol-13-acetate-induced MCF-7 human breast cancer cells. Arch Toxicol 2013; 87:857-69. [PMID: 23288142 DOI: 10.1007/s00204-012-1003-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 12/18/2012] [Indexed: 12/22/2022]
Abstract
Matrix metalloproteinase-9 (MMP-9) plays a crucial role in tumor metastasis. Previous studies showed that polyunsaturated fatty acids exhibit an anti-cancer effect in various human carcinoma cells, but the effect of docosahexaenoic acid (DHA) and linoleic acid (LA) on metastasis of breast cancer cells is not fully clarified. We studied the anti-metastasis potential of DHA and LA in 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced MCF-7 cells. We found that TPA (100 ng/ml) induced MMP-9 enzyme activity both dose- and time-dependently, and 200 μM DHA and LA significantly inhibited MMP-9 mRNA and protein expression, enzyme activity, cell migration, and invasion. Treatment with PD98059 (10 μM), wortmannin (10 μM), and GF109203X (0.5 μM) decreased TPA-induced MMP-9 protein expression and enzyme activity. TPA-induced activation of ERK1, Akt, and PKCδ was attenuated by DHA, whereas LA attenuated only ERK1 activation. GF109203X also suppressed ERK1 activation. EMSA showed that DHA, LA, PD98059, and wortmannin decreased TPA-induced NF-κB and AP-1 DNA-binding activity. Furthermore, DHA rather than LA dose-dependently increased HO-1 expression. HO-1 siRNA alleviated the inhibition by DHA of TPA-induced MMP-9 protein expression and enzyme activity in MCF-7 cells, and HO-1 knockdown reversed the DHA inhibition of cell migration. These results suggest that DHA and LA have both similar and divergent signaling pathways in the suppression of TPA-induced MCF-7 metastasis.
Collapse
Affiliation(s)
- Haw-Wen Chen
- Department of Nutrition, China Medical University, Taichung, Taiwan, ROC
| | | | | | | | | | | | | |
Collapse
|
44
|
Jeon WK, Hong HY, Seo WC, Lim KH, Lee HY, Kim WJ, Song SY, Kim BC. Smad7 sensitizes A549 lung cancer cells to cisplatin-induced apoptosis through heme oxygenase-1 inhibition. Biochem Biophys Res Commun 2012; 420:288-92. [PMID: 22421218 DOI: 10.1016/j.bbrc.2012.02.151] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2012] [Accepted: 02/28/2012] [Indexed: 01/10/2023]
Abstract
Smad7, an inhibitory Smad, acts as a key regulator forming autoinhibitory feedback loop in transforming growth factor-beta (TGF-β) signaling. However, a growing body of evidences suggests that Smad7 is capable of apoptotic function. In the present study, we have demonstrated a proapoptotic function of Smad7 as a negative regulator of survival protein heme oxygenase-1 (HO-1). The HO-1 protein level was elevated in cisplatin-resistant A549 human lung cancer cells and blockade of HO-1 activation sensitized the cells to apoptosis. Interestingly, overexpression of Smad7 decreased HO-1 gene expression and its enzymatic activity. Notably, Smad7 reduced Akt activity and infection with adenovirus expressing a constitutively active form of the Akt reversed the inhibitory effects of Smad7 to HO-1, indicating a negative action mechanism of Smad7 to Akt-HO-1-linked survival pathway. Consistently, Smad7 sensitized A549 cells to cisplatin-induced apoptosis and these effects were dependent on HO-1 and Akt inhibition. Based on these findings, we suggest that targeting Smad7 may be an efficient strategy for overcoming drug-resistance in cancer therapy.
Collapse
Affiliation(s)
- Woo-Kwang Jeon
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon 200-701, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Hishiki T, Yamamoto T, Morikawa T, Kubo A, Kajimura M, Suematsu M. Carbon monoxide: impact on remethylation/transsulfuration metabolism and its pathophysiologic implications. J Mol Med (Berl) 2012; 90:245-54. [PMID: 22331189 PMCID: PMC3296020 DOI: 10.1007/s00109-012-0875-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 01/27/2012] [Accepted: 01/31/2012] [Indexed: 01/08/2023]
Abstract
Carbon monoxide (CO) is a gaseous product generated by heme oxygenase (HO), which oxidatively degrades heme. While the stress-inducible HO-1 has well been recognized as an anti-oxidative defense mechanism under stress conditions, recent studies suggest that cancer cells utilize the reaction for their survival. HO-2, the constitutive isozyme, also plays protective roles as a tonic regulator for neurovascular function. Although protective roles of the enzyme reaction and CO have extensively been studied, little information is available on the molecular mechanisms by which the gas exerts its biological actions. Recent studies using metabolomics revealed that CO inhibits cystathionine β-synthase (CBS), which generates H2S, another gaseous mediator. The CO-dependent CBS inhibition may impact on the remethylation cycle and related metabolic pathways including the methionine salvage pathway and polyamine synthesis. This review focuses on the gas-responsive regulation of metabolic systems, particularly the remethylation and transsulfuration pathways, and their putative implications for cancer and ischemic diseases.
Collapse
Affiliation(s)
- Takako Hishiki
- Department of Biochemistry, JST, ERATO, Suematsu Gas Biology Project, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | |
Collapse
|