1
|
Moghadam D, Zarei R, Rostami A, Samare-Najaf M, Ghojoghi R, Savardashtaki A, Jafarinia M, Vakili S, Irajie C. The Growth Inhibitory Effect of Resveratrol and Gallic Acid on Prostate Cancer Cell Lines through the Alteration of Oxidative Stress Balance: The Interplay between Nrf2, HO-1, and BACH1 Genes. Anticancer Agents Med Chem 2024; 24:1220-1232. [PMID: 38984567 DOI: 10.2174/0118715206317999240708062744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/24/2024] [Accepted: 06/12/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND The association between oxidative stress and prostate cancer (PC) has been demonstrated both epidemiologically and experimentally. Balance in reactive oxygen species (ROS) levels depends on multiple factors, such as the expression of Nrf2, HO-1, and BACH1 genes. Natural polyphenols, such as resveratrol (RSV) and gallic acid (GA), affect cellular oxidative profiles. OBJECTIVE The present study investigated the possible effects of GA and RSV on the oxidative profiles of PC3 and DU145 cells, as well as Nrf2, HO-1, and BACH1 gene expression to achieve an understanding of the mechanisms involved. METHODS PC3 and DU145 cells were treated with ascending concentrations of RSV and GA for 72 h. Then cell growth and mRNA expression of Nrf2, HO-1, and BACH1 genes were analyzed by real-time PCR. Various spectrophotometric analyses were performed to measure oxidative stress markers. RESULTS RSV and GA significantly decreased the growth of PC3 and DU145 cells compared to the control group in a concentration-dependent manner. RSV and GA also decreased ROS production in PC3 cells, but in DU145 cells, only the latter polyphenol significantly decreased ROS content. In addition, RSV and GA had ameliorating effects on SOD, GR, GPX, and CAT activities and GSH levels in both cell lines. Also, RSV and GA induced HO- 1 and Nrf2 gene expression in both cell lines. BACH1 gene expression was induced by RSV only at lower concentrations, in contrast to GA in both cell lines. CONCLUSION Our data suggest that RSV and GA can prevent the growth of prostate cancer cells by disrupting oxidative stress-related pathways, such as changes in Nrf2, HO-1, and BACH1 gene expression.
Collapse
Affiliation(s)
- Delaram Moghadam
- Autophagy Research Center, Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Zarei
- Autophagy Research Center, Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amirabbas Rostami
- Department of Internal Medicine, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Samare-Najaf
- Autophagy Research Center, Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Rozita Ghojoghi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Morteza Jafarinia
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sina Vakili
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Cambyz Irajie
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
2
|
Oza PP, Kashfi K. The Triple Crown: NO, CO, and H 2S in cancer cell biology. Pharmacol Ther 2023; 249:108502. [PMID: 37517510 PMCID: PMC10529678 DOI: 10.1016/j.pharmthera.2023.108502] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/16/2023] [Accepted: 07/19/2023] [Indexed: 08/01/2023]
Abstract
Nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S) are three endogenously produced gases with important functions in the vasculature, immune defense, and inflammation. It is increasingly apparent that, far from working in isolation, these three exert many effects by modulating each other's activity. Each gas is produced by three enzymes, which have some tissue specificities and can also be non-enzymatically produced by redox reactions of various substrates. Both NO and CO share similar properties, such as activating soluble guanylate cyclase (sGC) to increase cyclic guanosine monophosphate (cGMP) levels. At the same time, H2S both inhibits phosphodiesterase 5A (PDE5A), an enzyme that metabolizes sGC and exerts redox regulation on sGC. The role of NO, CO, and H2S in the setting of cancer has been quite perplexing, as there is evidence for both tumor-promoting and pro-inflammatory effects and anti-tumor and anti-inflammatory activities. Each gasotransmitter has been found to have dual effects on different aspects of cancer biology, including cancer cell proliferation and apoptosis, invasion and metastasis, angiogenesis, and immunomodulation. These seemingly contradictory actions may relate to each gas having a dual effect dependent on its local flux. In this review, we discuss the major roles of NO, CO, and H2S in the context of cancer, with an effort to highlight the dual nature of each gas in different events occurring during cancer progression.
Collapse
Affiliation(s)
- Palak P Oza
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA; Graduate Program in Biology, City University of New York Graduate Center, New York 10091, USA.
| |
Collapse
|
3
|
Luh HT, Chen KW, Yang LY, Chen YT, Lin SH, Wang KC, Lai DM, Hsieh ST. Does a negative correlation of heme oxygenase-1 with hematoma thickness in chronic subdural hematomas affect neovascularization and microvascular leakage? A retrospective study with preliminary validation. J Neurosurg 2023; 139:536-543. [PMID: 36609367 DOI: 10.3171/2022.11.jns221790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/29/2022] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Chronic subdural hematoma (CSDH) is a common neurological disease among elderly adults. The progression of CSDH is an angiogenic process, involving inflammatory mediators that affect vascular permeability, microvascular leakage, and hematoma thickness. The authors aimed to identify biomarkers associated with angiogenesis and vascular permeability that might influence midline shift and hematoma thickness. METHODS Medical records and laboratory data of consecutive patients who underwent surgery for CSDH were analyzed. Collected data were basic demographic data, CSDH classification, CSDH thickness, midline shift, heme oxygenase-1 (HO-1) levels in hematomas, and common laboratory markers. Linear regression analysis was used to evaluate the relationship of CSDH thickness with characteristic variables. The chick chorioallantoic membrane (CAM) assay was used to test the angiogenic potency of identified variables in ex ovo culture of chick embryos. RESULTS In total, 93 patients with CSDH (71.0% male) with a mean age of 71.0 years were included. The mean CSDH thickness and midline shift were 19.7 and 9.8 mm, respectively. The mean levels of HO-1, ferritin, total bilirubin, white blood cells, segmented neutrophils, lymphocytes, platelets, international normalized ratio, and partial thromboplastin time were 36 ng/mL, 14.8 μg/mL, 10.5 mg/dL, 10.3 × 103 cells/μL, 69%, 21.7%, 221.1 × 109 cells/μL, 1.0, and 27.8 seconds, respectively. Pearson correlation analysis revealed that CSDH thickness was positively correlated with midline shift distance (r = 0.218, p < 0.05) but negatively correlated with HO-1 concentration (r = -0.364, p < 0.01) and ferritin level (r = -0.222, p < 0.05). Multivariate linear regression analysis revealed that HO-1 was an independent predictor of CSDH thickness (β = -0.084, p = 0.006). The angiogenic potency of HO-1 in hematoma fluid was tested with the chick CAM assay; topical addition of CSDH fluid with low HO-1 levels promoted neovascularization and microvascular leakage. Addition of HO-1 in a rescue experiment inhibited CSDH fluid-mediated angiogenesis and microvascular leakage. CONCLUSIONS HO-1 is an independent risk factor in CSDH hematomas and is negatively correlated with CSDH thickness. HO-1 may play a role in the pathophysiology and development of CSDH, possibly by preventing neovascularization and reducing capillary fragility and hyperpermeability.
Collapse
Affiliation(s)
- Hui-Tzung Luh
- 1Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- 2Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Kuo-Wei Chen
- 1Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- 2Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Ling-Yu Yang
- 3Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Tzu Chen
- 3Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Sheng-Hsuan Lin
- 4Institute of Statistics, National Yang Ming Chiao Tung University, Hsin-Chu, Taiwan
| | - Kuo-Chuan Wang
- 3Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Dar-Ming Lai
- 3Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Sung-Tsang Hsieh
- 5Department of Neurology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan; and
- 6Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
4
|
Ben-Eltriki M, Gayle EJ, Walker N, Deb S. Pharmacological Significance of Heme Oxygenase 1 in Prostate Cancer. Curr Issues Mol Biol 2023; 45:4301-4316. [PMID: 37232742 DOI: 10.3390/cimb45050273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
Heme oxygenase 1 (HO-1) is a detoxifying antioxidant microsomal enzyme that regulates inflammation, apoptosis, cell proliferation, and angiogenesis in prostate cancer (PCa). This makes HO-1 a promising target for therapeutic prevention and treatment due to its anti-inflammatory properties and ability to control redox homeostasis. Clinical evidence highlights the possible correlation between HO-1 expression and PCa growth, aggressiveness, metastasized tumors, resistance to therapy, and poor clinical outcomes. Interestingly, studies have reported anticancer benefits mediated by both HO-1 induction and inhibition in PCa models. Contrasting evidence exists on the role of HO-1 in PCa progression and possible treatment targets. Herein, we provide an overview of available evidence on the clinical significance of HO-1 signaling in PCa. It appears that the beneficial effects of HO-1 induction or inhibition are dependent on whether it is a normal versus malignant cell as well as the intensity (major vs. minor) of the increase in HO-1 enzymatic activity. The current literature evidence indicates that HO-1 has dual effects in PCa. The amount of cellular iron and reactive oxygen species (ROS) can determine the role of HO-1 in PCa. A major increase in ROS enforces HO-1 to a protective role. HO-1 overexpression may provide cryoprotection to normal cells against oxidative stress via suppressing the expression of proinflammatory genes, and thus offer therapeutic prevention. In contrast, a moderate increase in ROS can lead to the perpetrator role of HO-1, which is associated with PCa progression and metastasis. HO-1 inhibition by xenobiotics in DNA-damaged cells tilts the balance to promote apoptosis and inhibit PCa proliferation and metastasis. Overall, the totality of the evidence revealed that HO-1 may play a dual role in the therapeutic prevention and treatment of PCa.
Collapse
Affiliation(s)
- Mohamed Ben-Eltriki
- Department of Pharmacology and Therapeutics, Clinical Pharmacology Lab, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
- Cochrane Hypertension Review Group, Therapeutic Initiative, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Erysa J Gayle
- College of Biomedical Sciences, Larkin University, 18301 N. Miami Avenue, Miami, FL 33169, USA
| | - Noah Walker
- College of Biomedical Sciences, Larkin University, 18301 N. Miami Avenue, Miami, FL 33169, USA
| | - Subrata Deb
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL 33169, USA
| |
Collapse
|
5
|
Carbon Monoxide-Releasing Molecule-3 Suppresses the Malignant Biological Behavior of Tongue Squamous Cell Carcinoma via Regulating Keap1/Nrf2/HO-1 Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9418332. [PMID: 36158873 PMCID: PMC9507709 DOI: 10.1155/2022/9418332] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/05/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022]
Abstract
Carbon monoxide-releasing molecule-3 (CORM-3) is a water-soluble complex which has the ability to release carbon monoxide (CO). The study is aimed at investigating the epidemiological characters and effects of CORM-3 on tongue squamous cell carcinoma (TSCC) cells and the mechanisms involved. Firstly, CAL27 and SCC4 were treated with CORM-3 or iCORM-3. The proliferation, migration, and invasion of cells were separately evaluated by CCK-8, scratch assay, and transwell assay. We found that the optimal concentration of CORM-3 on the proliferation of CAL27 and SCC4 cells was 400 μM, and CORM-3 was significantly inhibited the proliferation, migration, and invasion of TSCC cells. Meanwhile, CORM-3 increased the protein expression of HO-1 detected by western blot. Short-hairpin RNAs (shRNAs) were constructed to manipulate the expression of HO-1 in CAL27 and SCC4 cells. Then, rescue assays were conducted to explore the reversion effect of shHO-1 on the CORM-3 function. Mechanistically, CORM-3 decreased the protein of Keap1 expression as well as increased Nrf2 expression. Upregulation of E-cadherin was observed, as well as the downregulation of N-cadherin expression significantly. The antitumor effect of CORM-3 was used to xenograft tumor in nude mice for further investigation in vivo, and the result showed that CORM-3 significantly suppressed tumor growth in xenograft nude mice. These data suggest that CORM-3 acts as a tumor suppressor by regulating the Keap1/Nrf2/HO-1 signaling pathway in TSCC, which provides a potential chemotherapeutic strategy for TSCC.
Collapse
|
6
|
Hsu SY, Wen ZH, Shih PC, Kuo HM, Lin SC, Liu HT, Lee YH, Wang YJ, Chen WF, Chen NF. Sinularin Induces Oxidative Stress-Mediated Apoptosis and Mitochondrial Dysfunction, and Inhibits Angiogenesis in Glioblastoma Cells. Antioxidants (Basel) 2022; 11:1433. [PMID: 35892635 PMCID: PMC9394238 DOI: 10.3390/antiox11081433] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 12/02/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a cancer of largely unknown cause that leads to a 5-year survival rate of approximately 7% in the United States. Current treatment strategies are not effective, indicating a strong need for the development of novel therapies. In this study, the outcomes of sinularin, a marine-derived product, were evaluated against GBM. Our cellular studies using GBM cells revealed that sinularin induces cell death. The measured half maximal inhibitory concentrations (IC50) values ranged from 30 to 6 μM at 24-72 h. Cell death was induced via the generation of ROS leading to mitochondria-mediated apoptosis. This was evidenced by annexin V/propidium iodine staining and an upregulation of cleaved forms of the pro-apoptotic proteins caspase 9, 3, and PARP, and supported by CellROXTM Green, MitoSOXTM Red, and CM-H2DCFDA staining methods. In addition, we observed a downregulation of the antioxidant enzymes SOD1/2 and thioredoxin. Upon treatment with sinularin at the ~IC50 concentration, mitochondrial respiration capacities were significantly reduced, as shown by measuring the oxygen consumption rates and enzymatic complexes of oxidative phosphorylation. Intriguingly, sinularin significantly inhibited indicators of angiogenesis such as vessel tube formation, cell migration, and cell mobility in human umbilical vein endothelial cells or the fusion cell line EA.Hy926. Lastly, in a transgenic zebrafish model, intersegmental vessel formation was also significantly inhibited by sinularin treatment. These findings indicate that sinularin exerts anti-brain cancer properties that include apoptosis induction but also antiangiogenesis.
Collapse
Affiliation(s)
- Shih-Yuan Hsu
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (S.-Y.H.); (Z.-H.W.); (P.-C.S.); (H.-M.K.); (Y.-H.L.); (Y.-J.W.); (W.-F.C.)
- Department of Neurosurgery, Pingtung Christian Hospital, Pingtung 90059, Taiwan
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (S.-Y.H.); (Z.-H.W.); (P.-C.S.); (H.-M.K.); (Y.-H.L.); (Y.-J.W.); (W.-F.C.)
| | - Po-Chang Shih
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (S.-Y.H.); (Z.-H.W.); (P.-C.S.); (H.-M.K.); (Y.-H.L.); (Y.-J.W.); (W.-F.C.)
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Hsiao-Mei Kuo
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (S.-Y.H.); (Z.-H.W.); (P.-C.S.); (H.-M.K.); (Y.-H.L.); (Y.-J.W.); (W.-F.C.)
- Center for Neuroscience, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Sung-Chun Lin
- Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung 90059, Taiwan;
| | - Hsin-Tzu Liu
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan;
| | - Yi-Hsin Lee
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (S.-Y.H.); (Z.-H.W.); (P.-C.S.); (H.-M.K.); (Y.-H.L.); (Y.-J.W.); (W.-F.C.)
| | - Yi-Jen Wang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (S.-Y.H.); (Z.-H.W.); (P.-C.S.); (H.-M.K.); (Y.-H.L.); (Y.-J.W.); (W.-F.C.)
| | - Wu-Fu Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (S.-Y.H.); (Z.-H.W.); (P.-C.S.); (H.-M.K.); (Y.-H.L.); (Y.-J.W.); (W.-F.C.)
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Nan-Fu Chen
- Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
- Center for General Education, Cheng Shiu University, Kaohsiung 833301, Taiwan
| |
Collapse
|
7
|
Yang Q, Wang W. The Nuclear Translocation of Heme Oxygenase-1 in Human Diseases. Front Cell Dev Biol 2022; 10:890186. [PMID: 35846361 PMCID: PMC9277552 DOI: 10.3389/fcell.2022.890186] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 06/10/2022] [Indexed: 12/30/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in the degradation of heme to generate carbon monoxide (CO), free iron and biliverdin, which could then be converted to bilirubin by biliverdin reductase. HO-1 exhibits cytoprotective effects of anti-apoptosis, anti-oxidation, and anti-inflammation via these byproducts generated during the above process. In the last few years, despite the canonical function of HO-1 and possible biological significance of its byproducts, a noncanonical function, through which HO-1 exhibits functions in diseases independent of its enzyme activity, also has been reported. In this review, the noncanonical functions of HO-1 and its translocation in other subcellular compartments are summarized. More importantly, we emphasize the critical role of HO-1 nuclear translocation in human diseases. Intriguingly, this translocation was linked to tumorigenesis and tumor progression in lung, prostate, head, and neck squamous cell carcinomas and chronic myeloid leukemia. Given the importance of HO-1 nuclear translocation in human diseases, nuclear HO-1 as a novel target might be attractive for the prevention and treatment of human diseases.
Collapse
Affiliation(s)
- Qing Yang
- Department of Breast Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenqian Wang
- Department of Plastic Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Wenqian Wang,
| |
Collapse
|
8
|
Zhu L, Xiong H, Huang X, Guyonnet V, Ma M, Chen X, Zheng Y, Wang L, Hu G. Identification and molecular mechanisms of novel antioxidant peptides from two sources of eggshell membrane hydrolysates showing cytoprotection against oxidative stress: A combined in silico and in vitro study. Food Res Int 2022; 157:111266. [DOI: 10.1016/j.foodres.2022.111266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/15/2022] [Accepted: 04/17/2022] [Indexed: 11/04/2022]
|
9
|
Jagadeesh ASV, Fang X, Kim SH, Guillen-Quispe YN, Zheng J, Surh YJ, Kim SJ. Non-canonical vs. Canonical Functions of Heme Oxygenase-1 in Cancer. J Cancer Prev 2022; 27:7-15. [PMID: 35419301 PMCID: PMC8984652 DOI: 10.15430/jcp.2022.27.1.7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 01/18/2023] Open
Abstract
Heme oxygenase-1 (HO-1) is a critical stress-responsive enzyme that has antioxidant and anti-inflammatory functions. HO-1 catalyzes heme degradation, which gives rise to the formation of carbon monoxide (CO), biliverdin, and iron. The upregulation of HO-1 under pathological conditions associated with cellular stress represents an important cytoprotective defense mechanism by virtue of the anti-oxidant properties of the bilirubin and the anti-inflammatory effect of the CO produced. The same mechanism is hijacked by premalignant and cancerous cells. In recent years, however, there has been accumulating evidence supporting that the upregulation of HO-1 promotes cancer progression, independently of its catalytic activity. Such non-canonical functions of HO-1 are associated with its interaction with other proteins, particularly transcription factors. HO-1 also undergoes post-translational modifications that influence its stability, functional activity, cellular translocation, etc. HO-1 is normally present in the endoplasmic reticulum, but distinct subcellular localizations, especially in the nucleus, are observed in multiple cancers. The nuclear HO-1 modulates the activation of various transcription factors, which does not appear to be mediated by carbon monoxide and iron. This commentary summarizes the non-canonical functions of HO-1 in the context of cancer growth and progression and underlying regulatory mechanisms.
Collapse
Affiliation(s)
| | - Xizhu Fang
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Seong Hoon Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Yanymee N. Guillen-Quispe
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Jie Zheng
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Young-Joon Surh
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Su-Jung Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| |
Collapse
|
10
|
Jagadeesh ASV, Fang X, Kim SH, Guillen-Quispe YN, Zheng J, Surh YJ, Kim SJ. Non-canonical vs. Canonical Functions of Heme Oxygenase-1 in Cancer. J Cancer Prev 2022. [PMID: 35419301 DOI: 10.15430/jcp.2022.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023] Open
Abstract
Heme oxygenase-1 (HO-1) is a critical stress-responsive enzyme that has antioxidant and anti-inflammatory functions. HO-1 catalyzes heme degradation, which gives rise to the formation of carbon monoxide (CO), biliverdin, and iron. The upregulation of HO-1 under pathological conditions associated with cellular stress represents an important cytoprotective defense mechanism by virtue of the anti-oxidant properties of the bilirubin and the anti-inflammatory effect of the CO produced. The same mechanism is hijacked by premalignant and cancerous cells. In recent years, however, there has been accumulating evidence supporting that the upregulation of HO-1 promotes cancer progression, independently of its catalytic activity. Such non-canonical functions of HO-1 are associated with its interaction with other proteins, particularly transcription factors. HO-1 also undergoes post-translational modifications that influence its stability, functional activity, cellular translocation, etc. HO-1 is normally present in the endoplasmic reticulum, but distinct subcellular localizations, especially in the nucleus, are observed in multiple cancers. The nuclear HO-1 modulates the activation of various transcription factors, which does not appear to be mediated by carbon monoxide and iron. This commentary summarizes the non-canonical functions of HO-1 in the context of cancer growth and progression and underlying regulatory mechanisms.
Collapse
Affiliation(s)
| | - Xizhu Fang
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Seong Hoon Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Yanymee N Guillen-Quispe
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Jie Zheng
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Young-Joon Surh
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Su-Jung Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| |
Collapse
|
11
|
Lage-Vickers S, Sanchis P, Bizzotto J, Toro A, Sabater A, Lavignolle R, Anselmino N, Labanca E, Paez A, Navone N, Valacco MP, Cotignola J, Vazquez E, Gueron G. Exploiting Interdata Relationships in Prostate Cancer Proteomes: Clinical Significance of HO-1 Interactors. Antioxidants (Basel) 2022; 11:antiox11020290. [PMID: 35204174 PMCID: PMC8868058 DOI: 10.3390/antiox11020290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 12/03/2022] Open
Abstract
Prostate cancer (PCa) cells display abnormal expression of proteins resulting in an augmented capacity to resist chemotherapy and colonize distant organs. We have previously shown the anti-tumoral role of heme oxygenase 1 (HO-1) in this disease. In this work, we undertook a mass spectrometry-based proteomics study to identify HO-1 molecular interactors that might collaborate with its modulatory function in PCa. Among the HO-1 interactors, we identified proteins with nuclear localization. Correlation analyses, using the PCa GSE70770 dataset, showed a significant and positive correlation between HMOX1 and 6 of those genes. Alternatively, HMOX1 and YWHAZ showed a negative correlation. Univariable analyses evidenced that high expression of HNRNPA2B1, HSPB1, NPM1, DDB1, HMGA1, ZC3HAV1, and HMOX1 was associated with increased relapse-free survival (RFS) in PCa patients. Further, PCa patients with high HSPB1/HMOX1, DDB1/HMOX1, and YWHAZ/HMOX1 showed a worse RFS compared with patients with lower ratios. Moreover, a decrease in RFS for patients with higher scores of this signature was observed using a prognostic risk score model. However, the only factor significantly associated with a higher risk of relapse was high YWHAZ. Multivariable analyses confirmed HSPB1, DDB1, and YWHAZ independence from PCa clinic-pathological parameters. In parallel, co-immunoprecipitation analysis in PCa cells ascertained HO-1/14-3-3ζ/δ (protein encoded by YWHAZ) interaction. Herein, we describe a novel protein interaction between HO-1 and 14-3-3ζ/δ in PCa and highlight these factors as potential therapeutic targets.
Collapse
Affiliation(s)
- Sofia Lage-Vickers
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (S.L.-V.); (P.S.); (J.B.); (A.T.); (A.S.); (R.L.); (A.P.); (M.P.V.); (J.C.); (E.V.)
- CONICET—Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Pablo Sanchis
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (S.L.-V.); (P.S.); (J.B.); (A.T.); (A.S.); (R.L.); (A.P.); (M.P.V.); (J.C.); (E.V.)
- CONICET—Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Juan Bizzotto
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (S.L.-V.); (P.S.); (J.B.); (A.T.); (A.S.); (R.L.); (A.P.); (M.P.V.); (J.C.); (E.V.)
- CONICET—Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Ayelen Toro
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (S.L.-V.); (P.S.); (J.B.); (A.T.); (A.S.); (R.L.); (A.P.); (M.P.V.); (J.C.); (E.V.)
- CONICET—Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Agustina Sabater
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (S.L.-V.); (P.S.); (J.B.); (A.T.); (A.S.); (R.L.); (A.P.); (M.P.V.); (J.C.); (E.V.)
- CONICET—Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Rosario Lavignolle
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (S.L.-V.); (P.S.); (J.B.); (A.T.); (A.S.); (R.L.); (A.P.); (M.P.V.); (J.C.); (E.V.)
- CONICET—Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Nicolas Anselmino
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.A.); (E.L.); (N.N.)
| | - Estefania Labanca
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.A.); (E.L.); (N.N.)
| | - Alejandra Paez
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (S.L.-V.); (P.S.); (J.B.); (A.T.); (A.S.); (R.L.); (A.P.); (M.P.V.); (J.C.); (E.V.)
- CONICET—Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Nora Navone
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.A.); (E.L.); (N.N.)
| | - Maria P. Valacco
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (S.L.-V.); (P.S.); (J.B.); (A.T.); (A.S.); (R.L.); (A.P.); (M.P.V.); (J.C.); (E.V.)
- CONICET—Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Javier Cotignola
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (S.L.-V.); (P.S.); (J.B.); (A.T.); (A.S.); (R.L.); (A.P.); (M.P.V.); (J.C.); (E.V.)
- CONICET—Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Elba Vazquez
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (S.L.-V.); (P.S.); (J.B.); (A.T.); (A.S.); (R.L.); (A.P.); (M.P.V.); (J.C.); (E.V.)
- CONICET—Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Geraldine Gueron
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (S.L.-V.); (P.S.); (J.B.); (A.T.); (A.S.); (R.L.); (A.P.); (M.P.V.); (J.C.); (E.V.)
- CONICET—Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
- Correspondence: ; Tel.: +54-9114-408-7796; Fax: +54-9114-788-5755
| |
Collapse
|
12
|
Wu J, Li S, Li C, Cui L, Ma J, Hui Y. The non-canonical effects of heme oxygenase-1, a classical fighter against oxidative stress. Redox Biol 2021; 47:102170. [PMID: 34688156 PMCID: PMC8577501 DOI: 10.1016/j.redox.2021.102170] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 08/29/2021] [Indexed: 12/30/2022] Open
Abstract
The role of heme oxygenase-1 in resisting oxidative stress and cell protection has always been a hot research topic. With the continuous deepening of research, in addition to directly regulating redox by catalyzing the degradation of heme, HO-1 protein also participates in the gene expression level in a great diversity of methods, thereby initiating cell defense. Particularly the non-canonical nuclear-localized HO-1 and HO-1 protein interactions play the role of a warrior against oxidative stress. Besides, HO-1 may be a promising marker for disease prediction and detection in many clinical trials. Especially for malignant diseases, there may be new advances in the treatment of HO-1 by regulating abnormal ROS and metabolic signaling. The purpose of this review is to systematically sort out and describe several aspects of research to facilitate further detailed mechanism research and clinical application promotion in the future. The different subcellular localizations ofHO-1 implies that it has special functions. Nuclear HO-1 plays an indispensable role in gene regulation and other aspects. The interactions between HO-1 and others provide the possibility to participate in vital physiological processes. HO-1 may become a potential disease assessment marker.
Collapse
Affiliation(s)
- Jiajia Wu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Siyu Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Cheng Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Liying Cui
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Jiajia Ma
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Yang Hui
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China; Basic Medical Institute of Heilongjiang Medical Science Academy, PR China; Translational Medicine Center of Northern China, PR China.
| |
Collapse
|
13
|
Wang T, Ashrafi A, Modareszadeh P, Deese AR, Chacon Castro MDC, Alemi PS, Zhang L. An Analysis of the Multifaceted Roles of Heme in the Pathogenesis of Cancer and Related Diseases. Cancers (Basel) 2021; 13:4142. [PMID: 34439295 PMCID: PMC8393563 DOI: 10.3390/cancers13164142] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/08/2021] [Accepted: 08/13/2021] [Indexed: 12/28/2022] Open
Abstract
Heme is an essential prosthetic group in proteins and enzymes involved in oxygen utilization and metabolism. Heme also plays versatile and fascinating roles in regulating fundamental biological processes, ranging from aerobic respiration to drug metabolism. Increasing experimental and epidemiological data have shown that altered heme homeostasis accelerates the development and progression of common diseases, including various cancers, diabetes, vascular diseases, and Alzheimer's disease. The effects of heme on the pathogenesis of these diseases may be mediated via its action on various cellular signaling and regulatory proteins, as well as its function in cellular bioenergetics, specifically, oxidative phosphorylation (OXPHOS). Elevated heme levels in cancer cells intensify OXPHOS, leading to higher ATP generation and fueling tumorigenic functions. In contrast, lowered heme levels in neurons may reduce OXPHOS, leading to defects in bioenergetics and causing neurological deficits. Further, heme has been shown to modulate the activities of diverse cellular proteins influencing disease pathogenesis. These include BTB and CNC homology 1 (BACH1), tumor suppressor P53 protein, progesterone receptor membrane component 1 protein (PGRMC1), cystathionine-β-synthase (CBS), soluble guanylate cyclase (sGC), and nitric oxide synthases (NOS). This review provides an in-depth analysis of heme function in influencing diverse molecular and cellular processes germane to disease pathogenesis and the modes by which heme modulates the activities of cellular proteins involved in the development of cancer and other common diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA; (T.W.); (A.A.); (P.M.); (A.R.D.); (M.D.C.C.C.); (P.S.A.)
| |
Collapse
|
14
|
Petrone MV, Toro A, Vazquez Echegaray C, Francia MG, Solari C, Cosentino MS, Vazquez E, Guberman A. The pluripotency transcription factor OCT4 represses heme oxygenase-1 gene expression. FEBS Lett 2021; 595:1949-1961. [PMID: 34056710 DOI: 10.1002/1873-3468.14138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/17/2021] [Accepted: 05/23/2021] [Indexed: 01/03/2023]
Abstract
In embryonic stem (ES) cells, oxidative stress control is crucial for genomic stability, self-renewal, and cell differentiation. Heme oxygenase-1 (HO-1) is a key player of the antioxidant system and is also involved in stem cell differentiation and pluripotency acquisition. We found that the HO-1 gene is expressed in ES cells and induced after promoting differentiation. Moreover, downregulation of the pluripotency transcription factor (TF) OCT4 increased HO-1 mRNA levels in ES cells, and analysis of ChIP-seq public data revealed that this TF binds to the HO-1 gene locus in pluripotent cells. Finally, ectopic expression of OCT4 in heterologous systems repressed a reporter carrying the HO-1 gene promoter and the endogenous gene. Hence, this work highlights the connection between pluripotency and redox homeostasis.
Collapse
Affiliation(s)
- María Victoria Petrone
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-UBA, Buenos Aires, Argentina
| | - Ayelén Toro
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-UBA, Buenos Aires, Argentina
| | - Camila Vazquez Echegaray
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-UBA, Buenos Aires, Argentina
| | - Marcos Gabriel Francia
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-UBA, Buenos Aires, Argentina
| | - Claudia Solari
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-UBA, Buenos Aires, Argentina
| | - Maria Soledad Cosentino
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-UBA, Buenos Aires, Argentina
| | - Elba Vazquez
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales Universidad de Buenos Aires, Buenos Aires, Argentina.,IQUIBICEN, CONICET-UBA, Buenos Aires, Argentina
| | - Alejandra Guberman
- IQUIBICEN, CONICET-UBA, Buenos Aires, Argentina.,Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales Universidad de Buenos Aires, Argentina
| |
Collapse
|
15
|
Cascardo F, Anselmino N, Páez A, Labanca E, Sanchis P, Antico-Arciuch V, Navone N, Gueron G, Vázquez E, Cotignola J. HO-1 Modulates Aerobic Glycolysis through LDH in Prostate Cancer Cells. Antioxidants (Basel) 2021; 10:966. [PMID: 34208670 PMCID: PMC8235201 DOI: 10.3390/antiox10060966] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 01/13/2023] Open
Abstract
Prostate cancer (PCa) is the second most diagnosed malignancy and the fifth leading cause of cancer associated death in men worldwide. Dysregulation of cellular energetics has become a hallmark of cancer, evidenced by numerous connections between signaling pathways that include oncoproteins and key metabolic enzymes. We previously showed that heme oxygenase 1 (HO-1), a cellular homeostatic regulator counteracting oxidative and inflammatory damage, exhibits anti-tumoral activity in PCa cells, inhibiting cell proliferation, migration, tumor growth and angiogenesis. The aim of this study was to assess the role of HO-1 on the metabolic signature of PCa. After HO-1 pharmacological induction with hemin, PC3 and C4-2B cells exhibited a significantly impaired cellular metabolic rate, reflected by glucose uptake, ATP production, lactate dehydrogenase (LDH) activity and extracellular lactate levels. Further, we undertook a bioinformatics approach to assess the clinical significance of LDHA, LDHB and HMOX1 in PCa, identifying that high LDHA or low LDHB expression was associated with reduced relapse free survival (RFS). Interestingly, the shortest RFS was observed for PCa patients with low HMOX1 and high LDHA, while an improved prognosis was observed for those with high HMOX1 and LDHB. Thus, HO-1 induction causes a shift in the cellular metabolic profile of PCa, leading to a less aggressive phenotype of the disease.
Collapse
Affiliation(s)
- Florencia Cascardo
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (F.C.); (P.S.); (V.A.-A.); (G.G.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Nicolás Anselmino
- Department of Genitourinary Medical Oncology, The David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.A.); (E.L.); (N.N.)
| | - Alejandra Páez
- Unidad de Transferencia Genética, Instituto de Oncología “Dr. Angel H. Roffo”, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1417DTB, Argentina;
| | - Estefanía Labanca
- Department of Genitourinary Medical Oncology, The David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.A.); (E.L.); (N.N.)
| | - Pablo Sanchis
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (F.C.); (P.S.); (V.A.-A.); (G.G.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Valeria Antico-Arciuch
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (F.C.); (P.S.); (V.A.-A.); (G.G.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Nora Navone
- Department of Genitourinary Medical Oncology, The David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.A.); (E.L.); (N.N.)
| | - Geraldine Gueron
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (F.C.); (P.S.); (V.A.-A.); (G.G.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Elba Vázquez
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (F.C.); (P.S.); (V.A.-A.); (G.G.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Javier Cotignola
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (F.C.); (P.S.); (V.A.-A.); (G.G.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| |
Collapse
|
16
|
Nitti M, Ivaldo C, Traverso N, Furfaro AL. Clinical Significance of Heme Oxygenase 1 in Tumor Progression. Antioxidants (Basel) 2021; 10:antiox10050789. [PMID: 34067625 PMCID: PMC8155918 DOI: 10.3390/antiox10050789] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/30/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023] Open
Abstract
Heme oxygenase 1 (HO-1) plays a key role in cell adaptation to stressors through the antioxidant, antiapoptotic, and anti-inflammatory properties of its metabolic products. For these reasons, in cancer cells, HO-1 can favor aggressiveness and resistance to therapies, leading to poor prognosis/outcome. Genetic polymorphisms of HO-1 promoter have been associated with an increased risk of cancer progression and a high degree of therapy failure. Moreover, evidence from cancer biopsies highlights the possible correlation between HO-1 expression, pathological features, and clinical outcome. Indeed, high levels of HO-1 in tumor specimens often correlate with reduced survival rates. Furthermore, HO-1 modulation has been proposed in order to improve the efficacy of antitumor therapies. However, contrasting evidence on the role of HO-1 in tumor biology has been reported. This review focuses on the role of HO-1 as a promising biomarker of cancer progression; understanding the correlation between HO-1 and clinical data might guide the therapeutic choice and improve the outcome of patients in terms of prognosis and life quality.
Collapse
|
17
|
Ma R, Shimura T, Yin C, Okugawa Y, Kitajima T, Koike Y, Okita Y, Ohi M, Uchida K, Goel A, Yao L, Zhang X, Toiyama Y. Antitumor effects of Andrographis via ferroptosis-associated genes in gastric cancer. Oncol Lett 2021; 22:523. [PMID: 34025790 DOI: 10.3892/ol.2021.12784] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 03/31/2021] [Indexed: 12/24/2022] Open
Abstract
The overall prognosis of advanced/metastatic gastric cancer (GC) remains poor despite the development of pharmacotherapy. Therefore, other treatment options, such as complementary and alternative medicine, should be considered to overcome this aggressive malignancy. Andrographis, which is a generally unharmful botanical compound, has gained increasing interest for its anticancer effects in multiple malignancies via the regulation of cancer progression-associated signaling pathways. In the present study, a series of in vitro experiments (cell proliferation, colony formation and apoptosis assays) was designed to elucidate the antitumor potential and mechanism of Andrographis in GC cells. The present study demonstrated that Andrographis exerted antitumor effects in GC cell lines (MKN74 and NUGC4) by inhibiting proliferation, reducing colony formation and enhancing apoptotic activity. Furthermore, it was demonstrated that the expression levels of the ferroptosis-associated genes heme oxygenase-1, glutamate-cysteine ligase catalytic and glutamate-cysteine ligase modifier were significantly upregulated after Andrographis treatment in both GC cell lines in reverse transcription-quantitative PCR experiments (P<0.05); this finding was further confirmed by immunoblotting assays (P<0.05). In conclusion, to the best of our knowledge, the present study was the first to demonstrate that Andrographis possessed antitumor properties by altering the expression levels of ferroptosis-associated genes, thereby providing novel insights into the potential of Andrographis as an adjunctive treatment option for patients with metastatic GC.
Collapse
Affiliation(s)
- Ruiya Ma
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.,Department of Colorectal Surgery, Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Tadanobu Shimura
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Chengzeng Yin
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yoshinaga Okugawa
- Department of Genomic Medicine, Mie University Hospital, Tsu, Mie 514-8507, Japan
| | - Takahito Kitajima
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yuhki Koike
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yoshiki Okita
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Masaki Ohi
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Keiichi Uchida
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91016, USA
| | - Li Yao
- Department of Surgery, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Xueming Zhang
- Department of Colorectal Surgery, Tangshan Gongren Hospital, Tangshan, Hebei 063000, P.R. China
| | - Yuji Toiyama
- Department of Gastrointestinal and Pediatric Surgery, Institute of Life Sciences, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| |
Collapse
|
18
|
Luu Hoang KN, Anstee JE, Arnold JN. The Diverse Roles of Heme Oxygenase-1 in Tumor Progression. Front Immunol 2021; 12:658315. [PMID: 33868304 PMCID: PMC8044534 DOI: 10.3389/fimmu.2021.658315] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/15/2021] [Indexed: 12/16/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is an inducible intracellular enzyme that is expressed in response to a variety of stimuli to degrade heme, which generates the biologically active catabolites carbon monoxide (CO), biliverdin and ferrous iron (Fe2+). HO-1 is expressed across a range of cancers and has been demonstrated to promote tumor progression through a variety of mechanisms. HO-1 can be expressed in a variety of cells within the tumor microenvironment (TME), including both the malignant tumor cells as well as stromal cell populations such as macrophages, dendritic cells and regulatory T-cells. Intrinsically to the cell, HO-1 activity provides antioxidant, anti-apoptotic and cytoprotective effects via its catabolites as well as clearing toxic intracellular heme. However, the catabolites of heme degradation can also diffuse outside of the cell to extrinsically modulate the wider TME, influencing cellular functionality and biological processes which promote tumor progression, such as facilitating angiogenesis and metastasis, as well as promoting anti-inflammation and immune suppression. Pharmacological inhibition of HO-1 has been demonstrated to be a promising therapeutic approach to promote anti-tumor immune responses and inhibit metastasis. However, these biological functions might be context, TME and cell type-dependent as there is also conflicting reports for HO-1 activity facilitating anti-tumoral processes. This review will consider our current understanding of the role of HO-1 in cancer progression and as a therapeutic target in cancer.
Collapse
Affiliation(s)
- Kim Ngan Luu Hoang
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Joanne E Anstee
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - James N Arnold
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| |
Collapse
|
19
|
Nuclear Localization of Heme Oxygenase-1 in Pathophysiological Conditions: Does It Explain the Dual Role in Cancer? Antioxidants (Basel) 2021; 10:antiox10010087. [PMID: 33440611 PMCID: PMC7826503 DOI: 10.3390/antiox10010087] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 12/12/2022] Open
Abstract
Heme Oxygenase-1 (HO-1) is a type II detoxifying enzyme that catalyzes the rate-limiting step in heme degradation leading to the formation of equimolar quantities of carbon monoxide (CO), free iron and biliverdin. HO-1 was originally shown to localize at the smooth endoplasmic reticulum membrane (sER), although increasing evidence demonstrates that the protein translocates to other subcellular compartments including the nucleus. The nuclear translocation occurs after proteolytic cleavage by proteases including signal peptide peptidase and some cysteine proteases. In addition, nuclear translocation has been demonstrated to be involved in several cellular processes leading to cancer progression, including induction of resistance to therapy and enhanced metastatic activity. In this review, we focus on nuclear HO-1 implication in pathophysiological conditions with special emphasis on malignant processes. We provide a brief background on the current understanding of the mechanisms underlying how HO-1 leaves the sER membrane and migrates to the nucleus, the circumstances under which it does so and, maybe the most important and unknown aspect, what the function of HO-1 in the nucleus is.
Collapse
|
20
|
|
21
|
Ortiz E, Sanchis P, Bizzotto J, Lage-Vickers S, Labanca E, Navone N, Cotignola J, Vazquez E, Gueron G. Myxovirus Resistance Protein 1 (MX1), a Novel HO-1 Interactor, Tilts the Balance of Endoplasmic Reticulum Stress towards Pro-Death Events in Prostate Cancer. Biomolecules 2020; 10:E1005. [PMID: 32640729 PMCID: PMC7407234 DOI: 10.3390/biom10071005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 11/21/2022] Open
Abstract
The inflammatory tumor microenvironment is a fertile niche accelerating prostate cancer (PCa). We have reported that heme-oxygenase (HO-1) had a strong anti-tumoral effect in PCa. We previously undertook an in-depth proteomics study to build the HO-1 interactome in PCa. In this work, we used a bioinformatics approach to address the biological significance of HO-1 interactors. Open-access PCa datasets were mined to address the clinical significance of the HO-1 interactome in human samples. HO-1 interactors were clustered into groups according to their expression profile in PCa patients. We focused on the myxovirus resistance gene (MX1) as: (1) it was significantly upregulated under HO-1 induction; (2) it was the most consistently downregulated gene in PCa vs. normal prostate; (3) its loss was associated with decreased relapse-free survival in PCa; and (4) there was a significant positive correlation between MX1 and HMOX1 in PCa patients. Further, MX1 was upregulated in response to endoplasmic reticulum stress (ERS), and this stress triggered apoptosis and autophagy in PCa cells. Strikingly, MX1 silencing reversed ERS. Altogether, we showcase MX1 as a novel HO-1 interactor and downstream target, associated with ERS in PCa and having a high impact in the clinical setting.
Collapse
Affiliation(s)
- Emiliano Ortiz
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (E.O.); (P.S.); (J.B.); (S.L.-V.); (J.C.); (E.V.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Pablo Sanchis
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (E.O.); (P.S.); (J.B.); (S.L.-V.); (J.C.); (E.V.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Juan Bizzotto
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (E.O.); (P.S.); (J.B.); (S.L.-V.); (J.C.); (E.V.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Sofia Lage-Vickers
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (E.O.); (P.S.); (J.B.); (S.L.-V.); (J.C.); (E.V.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Estefania Labanca
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (E.L.); (N.N.)
| | - Nora Navone
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (E.L.); (N.N.)
| | - Javier Cotignola
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (E.O.); (P.S.); (J.B.); (S.L.-V.); (J.C.); (E.V.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Elba Vazquez
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (E.O.); (P.S.); (J.B.); (S.L.-V.); (J.C.); (E.V.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| | - Geraldine Gueron
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (E.O.); (P.S.); (J.B.); (S.L.-V.); (J.C.); (E.V.)
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires C1428EGA, Argentina
| |
Collapse
|
22
|
HO-1 Interactors Involved in the Colonization of the Bone Niche: Role of ANXA2 in Prostate Cancer Progression. Biomolecules 2020; 10:biom10030467. [PMID: 32197509 PMCID: PMC7175266 DOI: 10.3390/biom10030467] [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: 01/30/2020] [Revised: 02/24/2020] [Accepted: 03/12/2020] [Indexed: 12/24/2022] Open
Abstract
Background: Prostate cancer (PCa) dissemination shows a tendency to develop in the bone, where heme oxygenase 1 (HO-1) plays a critical role in bone remodeling. Previously by LC/ESI-MSMS, we screened for HO-1 interacting proteins and identified annexin 2 (ANXA2). The aim of this study was to analyze the relevance of ANXA2/HO-1 in PCa and bone metastasis. Methods: We assessed ANXA2 levels using a co-culture transwell system of PC3 cells (pre-treated or not with hemin, an HO-1 specific inducer) and the pre-osteoclastic Raw264.7 cell line. Results: Under co-culture conditions, ANXA2 mRNA levels were significantly modulated in both cell lines. Immunofluorescence analysis unveiled a clear ANXA2 reduction in cell membrane immunostaining for Raw264.7 under the same conditions. This effect was supported by the detection of a decrease in Ca2+ concentration in the conditioned medium. HO-1 induction in tumor cells prevented both, the ANXA2 intracellular relocation and the decrease in Ca2+ concentration. Further, secretome analysis revealed urokinase (uPA) as a key player in the communication between osteoclast progenitors and PC3 cells. To assess the clinical significance of ANXA2/HO-1, we performed a bioinformatics analysis and identified that low expression of each gene strongly associated with poor prognosis in PCa regardless of the clinico-pathological parameters assessed. Further, these genes appear to behave in a dependent manner. Conclusions: ANXA2/HO-1 rises as a critical axis in PCa.
Collapse
|
23
|
Castruccio Castracani C, Longhitano L, Distefano A, Di Rosa M, Pittalà V, Lupo G, Caruso M, Corona D, Tibullo D, Li Volti G. Heme Oxygenase-1 and Carbon Monoxide Regulate Growth and Progression in Glioblastoma Cells. Mol Neurobiol 2020; 57:2436-2446. [PMID: 32108290 DOI: 10.1007/s12035-020-01869-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 01/06/2020] [Indexed: 01/11/2023]
Abstract
In human glioma tumours, heme oxygenase-1 (HO-1) is overexpressed when compared with normal brain tissues and during oligodendroglioma progression. However, the molecular mechanisms mediated by HO-1 to promote glioblastoma remain unknown. We therefore aimed at investigating the effect of HO-1 expression and its selective enzymatic inhibition in two different cell lines (i.e. A172 and U87-MG). HO-1 was induced by hemin treatment (10 μM), and VP13/47 (100 μM) was used as a specific non-competitive inhibitor of HO-1 activity. Cell proliferation was measured by cell index measurement (xCelligence technology) and clonogenic assay, whereas cell migration was assessed by wound healing assay. Carbon monoxide-releasing molecules (CORMs) (i.e. CORM-3 and CORM-A1) were also used in a separate set of experiments to confirm the effect of HO-1 by-product in glioblastoma progression further. Our results were further validated using GSE4412 microarray dataset analysis and comparing biopsies overexpressing HO-1 with the rest of the cases. Our results showed that hemin was able to induce both HO-1 gene and protein expression in a cell-dependent manner being A172 more responsive to pharmacological upregulation of HO-1. Hemin, but not CORMs treatment, resulted in a significant increase of cell proliferation following 24 h of treatment as measured by increased cell index and colony formation capacity and such effect was abolished by VP13/47. Interestingly, both hemin and CORMs showed a significant effect on the wound healing assay also exhibiting cell specificity. Finally, our dataset analysis showed a positive correlation of HO-1 gene expression with ITGBI and ITGBII which are membrane receptors involved in cell adhesion, embryogenesis, tissue repair, immune response and metastatic diffusion of tumour cells. In conclusion, our data suggest that HO-1 and its by-product CO exhibit a cell-specific effect on various aspects of disease progression and are associated with a complex series of molecular mechanisms driving cell proliferation, survival and metastasis.
Collapse
Affiliation(s)
- Carlo Castruccio Castracani
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95125, Catania, Italy
| | - Lucia Longhitano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95125, Catania, Italy
| | - Alfio Distefano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95125, Catania, Italy
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95125, Catania, Italy
| | - Valeria Pittalà
- Department of Drug Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Gabriella Lupo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95125, Catania, Italy
| | - Massimo Caruso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95125, Catania, Italy
| | - Daniela Corona
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95125, Catania, Italy
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95125, Catania, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95125, Catania, Italy.
| |
Collapse
|
24
|
Ho CJ, Gorski SM. Molecular Mechanisms Underlying Autophagy-Mediated Treatment Resistance in Cancer. Cancers (Basel) 2019; 11:E1775. [PMID: 31717997 PMCID: PMC6896088 DOI: 10.3390/cancers11111775] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 12/13/2022] Open
Abstract
Despite advances in diagnostic tools and therapeutic options, treatment resistance remains a challenge for many cancer patients. Recent studies have found evidence that autophagy, a cellular pathway that delivers cytoplasmic components to lysosomes for degradation and recycling, contributes to treatment resistance in different cancer types. A role for autophagy in resistance to chemotherapies and targeted therapies has been described based largely on associations with various signaling pathways, including MAPK and PI3K/AKT signaling. However, our current understanding of the molecular mechanisms underlying the role of autophagy in facilitating treatment resistance remains limited. Here we provide a comprehensive summary of the evidence linking autophagy to major signaling pathways in the context of treatment resistance and tumor progression, and then highlight recently emerged molecular mechanisms underlying autophagy and the p62/KEAP1/NRF2 and FOXO3A/PUMA axes in chemoresistance.
Collapse
Affiliation(s)
- Cally J. Ho
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 1L3, Canada;
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Sharon M. Gorski
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 1L3, Canada;
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| |
Collapse
|
25
|
Gandini NA, Alonso EN, Fermento ME, Mascaró M, Abba MC, Coló GP, Arévalo J, Ferronato MJ, Guevara JA, Núñez M, Pichel P, Curino AC, Facchinetti MM. Heme Oxygenase-1 Has an Antitumor Role in Breast Cancer. Antioxid Redox Signal 2019; 30:2030-2049. [PMID: 30484334 DOI: 10.1089/ars.2018.7554] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aims: Heme oxygenase-1 (HO-1) is an enzyme involved in cellular responses to oxidative stress and has also been shown to regulate processes related to cancer progression. In this regard, HO-1 has been shown to display a dual effect with either antitumor or protumor activity, which is also true for breast cancer (BC). In this work, we address this discrepancy regarding the role of HO-1 in BC. Results: HO-1 was detected in human BC tissues, and its protein levels correlated with reduced tumor size and longer overall survival time of patients, thus suggesting the clinical importance of HO-1 in this type of cancer. Contrariwise, nuclear localization of HO-1 correlated with higher tumor grade suggesting that the effect of HO-1 is dependent on its cellular localization. In vivo experiments showed that both pharmacological activation and genetic overexpression of HO-1 reduce the tumor burden in two different animal models of BC. Furthermore, the pharmacological and genetic activation of HO-1 in several BC cell lines reduce the cellular viability by inducing apoptosis and cell cycle arrest and decrease the cellular migration and invasion rates by modulating pathways involved in the epithelial-mesenchymal transition. Furthermore, HO-1 activation impaired in vivo the metastatic dissemination. Innovation and Conclusion: By using various BC cell lines and animal models as well as human tumor samples, we demonstrated that total HO-1 displays antitumor activities in BC. Furthermore, our study suggests that HO-1 subcellular localization may explain the differential effects observed for the protein in different tumor types.
Collapse
Affiliation(s)
- Norberto Ariel Gandini
- 1 Laboratorio de Biología del Cáncer, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Dpto. de Biología, Bioquímica y Farmacia (UNS), Bahía Blanca, Argentina
| | - Eliana Noelia Alonso
- 1 Laboratorio de Biología del Cáncer, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Dpto. de Biología, Bioquímica y Farmacia (UNS), Bahía Blanca, Argentina
| | - María Eugenia Fermento
- 1 Laboratorio de Biología del Cáncer, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Dpto. de Biología, Bioquímica y Farmacia (UNS), Bahía Blanca, Argentina
| | - Marilina Mascaró
- 1 Laboratorio de Biología del Cáncer, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Dpto. de Biología, Bioquímica y Farmacia (UNS), Bahía Blanca, Argentina
| | - Martín Carlos Abba
- 2 CINIBA, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Georgina Pamela Coló
- 1 Laboratorio de Biología del Cáncer, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Dpto. de Biología, Bioquímica y Farmacia (UNS), Bahía Blanca, Argentina
| | - Julián Arévalo
- 1 Laboratorio de Biología del Cáncer, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Dpto. de Biología, Bioquímica y Farmacia (UNS), Bahía Blanca, Argentina.,3 Servicio de Patología del Hospital Interzonal de Agudos Dr. José Penna, Bahía Blanca, Argentina
| | - María Julia Ferronato
- 1 Laboratorio de Biología del Cáncer, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Dpto. de Biología, Bioquímica y Farmacia (UNS), Bahía Blanca, Argentina
| | - Josefina Alejandra Guevara
- 1 Laboratorio de Biología del Cáncer, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Dpto. de Biología, Bioquímica y Farmacia (UNS), Bahía Blanca, Argentina
| | - Myriam Núñez
- 4 Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pamela Pichel
- 5 Hospital Municipal de Agudos Dr Leónidas Lucero, Bahía Blanca, Argentina
| | - Alejandro Carlos Curino
- 1 Laboratorio de Biología del Cáncer, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Dpto. de Biología, Bioquímica y Farmacia (UNS), Bahía Blanca, Argentina
| | - María Marta Facchinetti
- 1 Laboratorio de Biología del Cáncer, Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Universidad Nacional del Sur (UNS)-CONICET, Dpto. de Biología, Bioquímica y Farmacia (UNS), Bahía Blanca, Argentina
| |
Collapse
|
26
|
Heme Oxygenase 1 Impairs Glucocorticoid Receptor Activity in Prostate Cancer. Int J Mol Sci 2019; 20:ijms20051006. [PMID: 30813528 PMCID: PMC6429053 DOI: 10.3390/ijms20051006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/18/2019] [Accepted: 02/21/2019] [Indexed: 01/27/2023] Open
Abstract
Glucocorticoids are used during prostate cancer (PCa) treatment. However, they may also have the potential to drive castration resistant prostate cancer (CRPC) growth via the glucocorticoid receptor (GR). Given the association between inflammation and PCa, and the anti-inflammatory role of heme oxygenase 1 (HO-1), we aimed at identifying the molecular processes governed by the interaction between HO-1 and GR. PCa-derived cell lines were treated with Hemin, Dexamethasone (Dex), or both. We studied GR gene expression by RTqPCR, protein expression by Western Blot, transcriptional activity using reporter assays, and nuclear translocation by confocal microscopy. We also evaluated the expression of HO-1, FKBP51, and FKBP52 by Western Blot. Hemin pre-treatment reduced Dex-induced GR activity in PC3 cells. Protein levels of FKBP51, a cytoplasmic GR-binding immunophilin, were significantly increased in Hemin+Dex treated cells, possibly accounting for lower GR activity. We also evaluated these treatments in vivo using PC3 tumors growing as xenografts. We found non-significant differences in tumor growth among treatments. Immunohistochemistry analyses revealed strong nuclear GR staining in almost all groups. We did not observe HO-1 staining in tumor cells, but high HO-1 reactivity was detected in tumor infiltrating macrophages. Our results suggest an association and crossed modulation between HO-1 and GR pathways.
Collapse
|
27
|
Maamoun H, Benameur T, Pintus G, Munusamy S, Agouni A. Crosstalk Between Oxidative Stress and Endoplasmic Reticulum (ER) Stress in Endothelial Dysfunction and Aberrant Angiogenesis Associated With Diabetes: A Focus on the Protective Roles of Heme Oxygenase (HO)-1. Front Physiol 2019; 10:70. [PMID: 30804804 PMCID: PMC6378556 DOI: 10.3389/fphys.2019.00070] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 01/21/2019] [Indexed: 12/17/2022] Open
Abstract
Type-2 diabetes prevalence is continuing to rise worldwide due to physical inactivity and obesity epidemic. Diabetes and fluctuations of blood sugar are related to multiple micro- and macrovascular complications, that are attributed to oxidative stress, endoplasmic reticulum (ER) activation and inflammatory processes, which lead to endothelial dysfunction characterized, among other features, by reduced availability of nitric oxide (NO) and aberrant angiogenic capacity. Several enzymatic anti-oxidant and anti-inflammatory agents have been found to play protective roles against oxidative stress and its downstream signaling pathways. Of particular interest, heme oxygenase (HO) isoforms, specifically HO-1, have attracted much attention as major cytoprotective players in conditions associated with inflammation and oxidative stress. HO operates as a key rate-limiting enzyme in the process of degradation of the iron-containing molecule, heme, yielding the following byproducts: carbon monoxide (CO), iron, and biliverdin. Because HO-1 induction was linked to pro-oxidant states, it has been regarded as a marker of oxidative stress; however, accumulating evidence has established multiple cytoprotective roles of the enzyme in metabolic and cardiovascular disorders. The cytoprotective effects of HO-1 depend on several cellular mechanisms including the generation of bilirubin, an anti-oxidant molecule, from the degradation of heme; the induction of ferritin, a strong chelator of free iron; and the release of CO, that displays multiple anti-inflammatory and anti-apoptotic actions. The current review article describes the major molecular mechanisms contributing to endothelial dysfunction and altered angiogenesis in diabetes with a special focus on the interplay between oxidative stress and ER stress response. The review summarizes the key cytoprotective roles of HO-1 against hyperglycemia-induced endothelial dysfunction and aberrant angiogenesis and discusses the major underlying cellular mechanisms associated with its protective effects.
Collapse
Affiliation(s)
- Hatem Maamoun
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Tarek Benameur
- College of Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Gianfranco Pintus
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| | - Shankar Munusamy
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Drake University, Des Moines, IA, United States
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, Qatar University, Doha, Qatar
| |
Collapse
|
28
|
Alsaad AM, Al-Arifi MN, Maayah ZH, Attafi IM, Alanazi FE, Belali OM, Alhoshani A, Asiri YA, Korashy HM. Genotoxic impact of long-term cigarette and waterpipe smoking on DNA damage and oxidative stress in healthy subjects. Toxicol Mech Methods 2018; 29:119-127. [DOI: 10.1080/15376516.2018.1528650] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Abdulaziz M. Alsaad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed N. Al-Arifi
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Zaid H. Maayah
- Cardiovascular Research Centre, Department of Pediatrics and Medicine, Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Ibraheem M. Attafi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Fawaz E. Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Osamah M. Belali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ali Alhoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Yousif A. Asiri
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hesham M. Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, Qatar University, Doha, Qatar
| |
Collapse
|
29
|
Jin H, Kim HS, Seo GS, Lee SH. A new chalcone derivative, 3-phenyl-1-(2,4,6-tris(methoxymethoxy)phenyl)prop-2-yn-1-one), inhibits phorbol ester-induced metastatic activity of colorectal cancer cells through upregulation of heme oxygenase-1. Eur J Pharmacol 2018; 841:1-9. [PMID: 30321531 DOI: 10.1016/j.ejphar.2018.10.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 10/10/2018] [Accepted: 10/10/2018] [Indexed: 12/22/2022]
Abstract
Chalcone (1,3-diphenyl-2-propen-1-one) derivatives exert anti-cancer activity by targeting key molecules that can lead to carcinogenesis. We synthesized the chalcone derivative 3-phenyl-1-(2,4,6-tris(methoxymethoxy)phenyl)prop-2-yn-1-one (KB-34) and previously reported its anti-inflammatory activity in macrophages. In this study, we examined the anti-metastatic activity of KB-34 against human colorectal cancer (CRC) cells and elucidated its underlying molecular mechanisms. KB-34 treatment significantly inhibited 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced migration, as well as the invasion and proliferation of CRC cells (HT-29 and SW620). TPA-induced activation of NF-κB was also markedly suppressed by KB-34 in HT-29 cells. KB-34 suppressed the expression of matrix metalloproteinase-7 (MMP-7) at both the mRNA and protein levels in TPA-stimulated CRC cells (HT-29 and SW620). We also demonstrated that induced heme oxygenase-1 (HO-1) expression in CRC cells (HT-29 and SW620) and HO-1 is required for KB-34-mediated suppression of the expression of MMP-7 in TPA-stimulated HT-29 cells. Additionally, the cyclin-dependent kinase inhibitor p21 was significantly induced by treatment with KB-34 in CRC cells (HT-29 and SW620). Knockdown of HO-1 prevented the induction of p21 expression by KB-34 in HT-29 cells. Furthermore, we also demonstrated that 5-fluorouracil (5-FU) together with KB-34 produced a significantly greater inhibition of growth and stimulation of apoptosis of HT-29 cells than did 5-FU alone. In conclusion, KB-34 inhibits the TPA-stimulated metastatic potential of HT-29 cells by induction of HO-1 and may be a promising anti-cancer agent in chemotherapeutic strategies for CRC.
Collapse
Affiliation(s)
- Hao Jin
- Institute of Pharmaceutical Research and Development, College of Pharmacy, Wonkwang University, Jeonbuk 54538, Republic of Korea
| | - Hak Sung Kim
- Institute of Pharmaceutical Research and Development, College of Pharmacy, Wonkwang University, Jeonbuk 54538, Republic of Korea
| | - Geom Seog Seo
- Digestive Disease Research Institute, Wonkwang University College of Medicine, Jeonbuk 54538, Republic of Korea
| | - Sung Hee Lee
- Institute of Pharmaceutical Research and Development, College of Pharmacy, Wonkwang University, Jeonbuk 54538, Republic of Korea.
| |
Collapse
|
30
|
Tsai CF, Chen JH, Chang CN, Lu DY, Chang PC, Wang SL, Yeh WL. Fisetin inhibits cell migration via inducing HO-1 and reducing MMPs expression in breast cancer cell lines. Food Chem Toxicol 2018; 120:528-535. [DOI: 10.1016/j.fct.2018.07.059] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 07/16/2018] [Accepted: 07/30/2018] [Indexed: 01/09/2023]
|
31
|
Herroon MK, Rajagurubandara E, Diedrich JD, Heath EI, Podgorski I. Adipocyte-activated oxidative and ER stress pathways promote tumor survival in bone via upregulation of Heme Oxygenase 1 and Survivin. Sci Rep 2018; 8:40. [PMID: 29311669 PMCID: PMC5758829 DOI: 10.1038/s41598-017-17800-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/30/2017] [Indexed: 01/26/2023] Open
Abstract
Metastatic tumor cells engage the local tumor microenvironment and activate specific pro-survival mechanisms to thrive and progress in the harsh bone marrow niche. Here we show that the major contributors to the survival of carcinoma cells that have colonized the bone marrow are the adipocyte-induced oxidative stress and ER stress pathways. We demonstrate that upon exposure to adipocyte-rich environments in vitro or in vivo, bone-trophic prostate and breast tumor cells upregulate the oxidative stress enzyme, HO-1. We also show that HO-1 levels are significantly increased in human metastatic prostate cancer tissues and that stable HO-1 overexpression in tumor cells promotes growth and invasiveness. Co-incident with the adipocyte-induced expression of HO-1, there is an upregulation of ER chaperone BIP and splicing of XBP1, indicating adipocyte-driven unfolded protein response, a process that we show to be sensitive to antioxidant treatment. Importantly, we also demonstrate that triggering of the oxidative stress and ER stress responses, or HO-1 induction by adipocyte exposure result in the activation of pro-survival pathways, involving survivin. Collectively, our findings reveal a new link between HO-1 and survivin expression in tumor cells, and provide a new insight into potentially targetable survival pathways in bone-metastatic disease.
Collapse
Affiliation(s)
- Mackenzie K Herroon
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, USA
| | | | - Jonathan D Diedrich
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, USA
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Elisabeth I Heath
- Oncology, Wayne State University School of Medicine, Detroit, MI, USA
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Izabela Podgorski
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, USA.
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
32
|
Jeddi F, Soozangar N, Sadeghi MR, Somi MH, Samadi N. Contradictory roles of Nrf2/Keap1 signaling pathway in cancer prevention/promotion and chemoresistance. DNA Repair (Amst) 2017; 54:13-21. [DOI: 10.1016/j.dnarep.2017.03.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 03/25/2017] [Accepted: 03/26/2017] [Indexed: 12/17/2022]
|
33
|
Jaworski FM, Gentilini LD, Gueron G, Meiss RP, Ortiz EG, Berguer PM, Ahmed A, Navone N, Rabinovich GA, Compagno D, Laderach DJ, Vazquez ES. In Vivo Hemin Conditioning Targets the Vascular and Immunologic Compartments and Restrains Prostate Tumor Development. Clin Cancer Res 2017; 23:5135-5148. [PMID: 28512172 DOI: 10.1158/1078-0432.ccr-17-0112] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 04/01/2017] [Accepted: 05/10/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Conditioning strategies constitute a relatively unexplored and exciting opportunity to shape tumor fate by targeting the tumor microenvironment. In this study, we assessed how hemin, a pharmacologic inducer of heme oxygenase-1 (HO-1), has an impact on prostate cancer development in an in vivo conditioning model.Experimental Design: The stroma of C57BL/6 mice was conditioned by subcutaneous administration of hemin prior to TRAMP-C1 tumor challenge. Complementary in vitro and in vivo assays were performed to evaluate hemin effect on both angiogenesis and the immune response. To gain clinical insight, we used prostate cancer patient-derived samples in our studies to assess the expression of HO-1 and other relevant genes.Results: Conditioning resulted in increased tumor latency and decreased initial growth rate. Histologic analysis of tumors grown in conditioned mice revealed impaired vascularization. Hemin-treated human umbilical vein endothelial cells (HUVEC) exhibited decreased tubulogenesis in vitro only in the presence of TRAMP-C1-conditioned media. Subcutaneous hemin conditioning hindered tumor-associated neovascularization in an in vivo Matrigel plug assay. In addition, hemin boosted CD8+ T-cell proliferation and degranulation in vitro and antigen-specific cytotoxicity in vivo A significant systemic increase in CD8+ T-cell frequency was observed in preconditioned tumor-bearing mice. Tumors from hemin-conditioned mice showed reduced expression of galectin-1 (Gal-1), key modulator of tumor angiogenesis and immunity, evidencing persistent remodeling of the microenvironment. We also found a subset of prostate cancer patient-derived xenografts and prostate cancer patient samples with mild HO-1 and low Gal-1 expression levels.Conclusions: These results highlight a novel function of a human-used drug as a means of boosting the antitumor response. Clin Cancer Res; 23(17); 5135-48. ©2017 AACR.
Collapse
Affiliation(s)
- Felipe M Jaworski
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales (FCEN), Departamento de Química Biológica (QB), Laboratorio de Inflamación y Cáncer, Buenos Aires, Argentina.,Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales (FCEN), Departamento de Química Biológica (QB), Laboratorio de Glico-Oncología Molecular y Funcional, Buenos Aires, Argentina.,CONICET - Universidad de Buenos Aires (UBA), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Lucas D Gentilini
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales (FCEN), Departamento de Química Biológica (QB), Laboratorio de Glico-Oncología Molecular y Funcional, Buenos Aires, Argentina.,CONICET - Universidad de Buenos Aires (UBA), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Geraldine Gueron
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales (FCEN), Departamento de Química Biológica (QB), Laboratorio de Inflamación y Cáncer, Buenos Aires, Argentina.,CONICET - Universidad de Buenos Aires (UBA), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Roberto P Meiss
- Department of Pathology, Institute of Oncological Studies, National Academy of Medicine, Buenos Aires, Argentina
| | - Emiliano G Ortiz
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales (FCEN), Departamento de Química Biológica (QB), Laboratorio de Inflamación y Cáncer, Buenos Aires, Argentina.,CONICET - Universidad de Buenos Aires (UBA), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Paula M Berguer
- Fundación Instituto Leloir (FIL) - IIBBA - CONICET, Buenos Aires, Argentina
| | - Asif Ahmed
- Aston Medical Research Institute, Aston Medical School, University of Aston, Birmingham, United Kingdom
| | - Nora Navone
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriel A Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina.,Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
| | - Daniel Compagno
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales (FCEN), Departamento de Química Biológica (QB), Laboratorio de Glico-Oncología Molecular y Funcional, Buenos Aires, Argentina.,CONICET - Universidad de Buenos Aires (UBA), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Diego J Laderach
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales (FCEN), Departamento de Química Biológica (QB), Laboratorio de Glico-Oncología Molecular y Funcional, Buenos Aires, Argentina. .,CONICET - Universidad de Buenos Aires (UBA), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina.,Departamento de Ciencias Básicas, Universidad Nacional de Luján, Buenos Aires, Argentina
| | - Elba S Vazquez
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales (FCEN), Departamento de Química Biológica (QB), Laboratorio de Inflamación y Cáncer, Buenos Aires, Argentina. .,CONICET - Universidad de Buenos Aires (UBA), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| |
Collapse
|
34
|
Nitti M, Piras S, Marinari UM, Moretta L, Pronzato MA, Furfaro AL. HO-1 Induction in Cancer Progression: A Matter of Cell Adaptation. Antioxidants (Basel) 2017; 6:antiox6020029. [PMID: 28475131 PMCID: PMC5488009 DOI: 10.3390/antiox6020029] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 04/26/2017] [Accepted: 04/29/2017] [Indexed: 02/07/2023] Open
Abstract
The upregulation of heme oxygenase-1 (HO-1) is one of the most important mechanisms of cell adaptation to stress. Indeed, the redox sensitive transcription factor Nrf2 is the pivotal regulator of HO-1 induction. Through the antioxidant, antiapoptotic, and antinflammatory properties of its metabolic products, HO-1 plays a key role in healthy cells in maintaining redox homeostasis and in preventing carcinogenesis. Nevertheless, several lines of evidence have highlighted the role of HO-1 in cancer progression and its expression correlates with tumor growth, aggressiveness, metastatic and angiogenetic potential, resistance to therapy, tumor escape, and poor prognosis, even though a tumor- and tissue-specific activity has been observed. In this review, we summarize the current literature regarding the pro-tumorigenic role of HO-1 dependent tumor progression as a promising target in anticancer strategy.
Collapse
Affiliation(s)
- Mariapaola Nitti
- Department of Experimental Medicine, University of Genoa, Via L. B. Alberti 2, Genoa 16132, Italy.
| | - Sabrina Piras
- Department of Experimental Medicine, University of Genoa, Via L. B. Alberti 2, Genoa 16132, Italy.
| | - Umberto M Marinari
- Department of Experimental Medicine, University of Genoa, Via L. B. Alberti 2, Genoa 16132, Italy.
| | - Lorenzo Moretta
- Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, Rome 00165, Italy.
| | - Maria A Pronzato
- Department of Experimental Medicine, University of Genoa, Via L. B. Alberti 2, Genoa 16132, Italy.
| | - Anna Lisa Furfaro
- Giannina Gaslini Institute, IRCCS, Via Gerolamo Gaslini 5, Genoa 16147, Italy.
| |
Collapse
|
35
|
Otterbein LE, Foresti R, Motterlini R. Heme Oxygenase-1 and Carbon Monoxide in the Heart: The Balancing Act Between Danger Signaling and Pro-Survival. Circ Res 2017; 118:1940-1959. [PMID: 27283533 DOI: 10.1161/circresaha.116.306588] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/02/2016] [Indexed: 12/22/2022]
Abstract
Understanding the processes governing the ability of the heart to repair and regenerate after injury is crucial for developing translational medical solutions. New avenues of exploration include cardiac cell therapy and cellular reprogramming targeting cell death and regeneration. An attractive possibility is the exploitation of cytoprotective genes that exist solely for self-preservation processes and serve to promote and support cell survival. Although the antioxidant and heat-shock proteins are included in this category, one enzyme that has received a great deal of attention as a master protective sentinel is heme oxygenase-1 (HO-1), the rate-limiting step in the catabolism of heme into the bioactive signaling molecules carbon monoxide, biliverdin, and iron. The remarkable cardioprotective effects ascribed to heme oxygenase-1 are best evidenced by its ability to regulate inflammatory processes, cellular signaling, and mitochondrial function ultimately mitigating myocardial tissue injury and the progression of vascular-proliferative disease. We discuss here new insights into the role of heme oxygenase-1 and heme on cardiovascular health, and importantly, how they might be leveraged to promote heart repair after injury.
Collapse
Affiliation(s)
- Leo E Otterbein
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Roberta Foresti
- Inserm, U955, Equipe 12, Créteil, 94000, France.,University Paris Est, Faculty of Medicine, Créteil, 94000, France
| | - Roberto Motterlini
- Inserm, U955, Equipe 12, Créteil, 94000, France.,University Paris Est, Faculty of Medicine, Créteil, 94000, France
| |
Collapse
|
36
|
Aspects of Carbon Monoxide in Form of CO-Releasing Molecules Used in Cancer Treatment: More Light on the Way. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9326454. [PMID: 28286606 PMCID: PMC5327762 DOI: 10.1155/2017/9326454] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 01/10/2017] [Accepted: 01/15/2017] [Indexed: 01/23/2023]
Abstract
Carbon monoxide (CO) has always been recognised as a toxic gas, due to its higher affinity for haemoglobin than oxygen. However, biological studies have revealed an intriguing role for CO as an endogenous signalling molecule, a gasotransmitter. CO is demonstrated to exert many cellular activities including anti-inflammatory, antiapoptotic, and antiproliferative activities. In animal studies, CO gas administration can prevent tissues from hypoxia or ischemic-reperfusion injury. As a result, there are a plethora of reports dealing with the biological applications of CO and CO-releasing molecules (CORMs) in inflammatory and vascular diseases. CORMs have already been tested as a therapeutic agent in clinical trials. More recently, an increased interest has been drawn to CO's potential use as an anticancer agent. In this review, we will aim to give an overview of the research focused on the role of CO and CORMs in different types of cancer and expand to the recent development of the next generation CORMs for clinical application in cancer treatment.
Collapse
|
37
|
Korashy HM, Attafi IM, Famulski KS, Bakheet SA, Hafez MM, Alsaad AMS, Al-Ghadeer ARM. Gene expression profiling to identify the toxicities and potentially relevant human disease outcomes associated with environmental heavy metal exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 221:64-74. [PMID: 27916491 DOI: 10.1016/j.envpol.2016.10.058] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 10/09/2016] [Accepted: 10/19/2016] [Indexed: 06/06/2023]
Abstract
Heavy metals are the most commonly encountered toxic substances that increase susceptibility to various diseases after prolonged exposure. We have previously shown that healthy volunteers living near a mining area had significant contamination with heavy metals associated with significant changes in the expression of some detoxifying genes, xenobiotic metabolizing enzymes, and DNA repair genes. However, alterations of most of the molecular target genes associated with diseases are still unknown. Thus, the aims of this study were to (a) evaluate the gene expression profile and (b) identify the toxicities and potentially relevant human disease outcomes associated with long-term human exposure to environmental heavy metals in mining area using microarray analysis. For this purpose, 40 healthy male volunteers who were residents of a heavy metal-polluted area (Mahd Al-Dhahab city, Saudi Arabia) and 20 healthy male volunteers who were residents of a non-heavy metal-polluted area were included in the study. Total RNA was isolated from whole blood using PAXgene Blood RNA tubes and then reversed transcribed and hybridized to the gene array using the Affymetrix U219 GeneChip. Microarray analysis showed about 2129 genes were identified and differentially altered, among which a shared set of 425 genes was differentially expressed in the heavy metal-exposed groups. Ingenuity pathway analysis revealed that the most altered gene-regulated diseases in heavy metal-exposed groups included hematological and developmental disorders and mostly renal and urological diseases. Quantitative real-time polymerase chain reaction closely matched the microarray data for some genes tested. Importantly, changes in gene-related diseases were attributed to alterations in the genes encoded for protein synthesis. Renal and urological diseases were the diseases that were most frequently associated with the heavy metal-exposed group. Therefore, there is a need for further studies to validate these genes, which could be used as early biomarkers to prevent renal injury.
Collapse
Affiliation(s)
- Hesham M Korashy
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia.
| | - Ibraheem M Attafi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Konrad S Famulski
- Alberta Transplant Applied Genomics Centre, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Saleh A Bakheet
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohammed M Hafez
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Abdulaziz M S Alsaad
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Abdul Rahman M Al-Ghadeer
- Central Laboratory, Research Center, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
38
|
Heme oxygenase-1 in the forefront of a multi-molecular network that governs cell-cell contacts and filopodia-induced zippering in prostate cancer. Cell Death Dis 2016; 7:e2570. [PMID: 28032857 PMCID: PMC5261017 DOI: 10.1038/cddis.2016.420] [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: 10/24/2016] [Accepted: 11/11/2016] [Indexed: 01/02/2023]
Abstract
Prostate cancer (PCa) cells display abnormal expression of cytoskeletal proteins resulting in an augmented capacity to resist chemotherapy and colonize distant organs. We have previously shown that heme oxygenase 1 (HO-1) is implicated in cell morphology regulation in PCa. Here, through a multi 'omics' approach we define the HO-1 interactome in PCa, identifying HO-1 molecular partners associated with the integrity of the cellular cytoskeleton. The bioinformatics screening for these cytoskeletal-related partners reveal that they are highly misregulated in prostate adenocarcinoma compared with normal prostate tissue. Under HO-1 induction, PCa cells present reduced frequency in migration events, trajectory and cell velocity and, a significant higher proportion of filopodia-like protrusions favoring zippering among neighboring cells. Moreover forced expression of HO-1 was also capable of altering cell protrusions in transwell co-culture systems of PCa cells with MC3T3 cells (pre-osteoblastic cell line). Accordingly, these effects were reversed under siHO. Transcriptomics profiling evidenced significant modulation of key markers related to cell adhesion and cell–cell communication under HO-1 induction. The integration from our omics-based research provides a four molecular pathway foundation (ANXA2/HMGA1/POU3F1; NFRSF13/GSN; TMOD3/RAI14/VWF; and PLAT/PLAU) behind HO-1 regulation of tumor cytoskeletal cell compartments. The complementary proteomics and transcriptomics approaches presented here promise to move us closer to unravel the molecular framework underpinning HO-1 involvement in the modulation of cytoskeleton pathways, pushing toward a less aggressive phenotype in PCa.
Collapse
|
39
|
Seo K, Ki SH, Park EY, Shin SM. 5-Fluorouracil inhibits cell migration by induction of Sestrin2 in colon cancer cells. Arch Pharm Res 2016; 40:231-239. [PMID: 28028695 DOI: 10.1007/s12272-016-0878-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 12/17/2016] [Indexed: 01/04/2023]
Abstract
5-Fluorouracil (5-FU) is a chemotherapeutic agent used in the treatment of colorectal cancer. In this study, we investigated whether 5-FU induces Sestrin2 (SESN2), an antioxidant enzyme, and the role of SESN2 in 5-FU action in colon cancer cells. We found that 5-FU upregulated SESN2 protein expression in both HCT116 and HT29 cells. It also increased transcripts of SESN1 and SESN2, but not of SESN3. Furthermore, we investigated whether production of reactive oxygen species (ROS) was involved in 5-FU-induced SESN2 expression. 5-FU did not increase ROS production nor affect Nrf2 phosphorylation and expression levels. Moreover, SESN2 upregulation by 5-FU was not prevented by pretreatment with antioxidants. Next, we investigated p53 levels after 5-FU treatment to elucidate the regulation of SESN2 by 5-FU. An increase in p53 levels was detected following 5-FU treatment; pifithrin-α, an inhibitor of p53 activation, reversed 5-FU-induced SESN2 expression. 5-FU prevented serum-induced in vitro cell migration, but knockdown of SESN2 or treatment with pifithrin-α reversed a 5-FU-mediated decrease in cell migration. Taken together, our results suggest that 5-FU increases SESN2 levels via a p53-dependent pathway, which contributes to inhibition of cancer cell migration in vitro.
Collapse
Affiliation(s)
- Kyuhwa Seo
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju, 501-759, South Korea
| | - Sung Hwan Ki
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju, 501-759, South Korea
| | - Eun Young Park
- College of Pharmacy, Mokpo National University, Muan-gun, Jeonnam, 534-729, South Korea
| | - Sang Mi Shin
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju, 501-759, South Korea.
| |
Collapse
|
40
|
Matsuo T, Miyata Y, Mitsunari K, Yasuda T, Ohba K, Sakai H. Pathological significance and prognostic implications of heme oxygenase 1 expression in non-muscle-invasive bladder cancer: Correlation with cell proliferation, angiogenesis, lymphangiogenesis and expression of VEGFs and COX-2. Oncol Lett 2016; 13:275-280. [PMID: 28123555 DOI: 10.3892/ol.2016.5416] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/03/2016] [Indexed: 01/09/2023] Open
Abstract
Heme oxygenase 1 (HO-1) is a stress-response protein and its expression is associated with malignant potential and poor prognosis in several types of cancer. The present study investigated the association between HO-1 expression levels and the pathological features, clinical outcomes and other associated factors in patients with non-muscle-invasive bladder cancer (NMIBC). HO-1 expression was evaluated using immunohistochemistry in 147 formalin-fixed tissue specimens. The proliferation index, microvessel density, lymph vessel density and expression of cyclooxygenase (COX)-2 and vascular endothelial growth factor (VEGF)-A, -C, and -D were also investigated. Correlations among variables were analyzed by multivariate analysis. Survival was assessed using Kaplan-Meier survival curves and multivariate statistics. HO-1 expression levels in high-grade and pT1 tumors were significantly higher compared with low-grade and pTa tumors, and were correlated with the proliferation index (P<0.001), lymph vessel density (P=0.021) and COX-2 expression levels (P=0.003). The proliferation index and COX-2 expression levels were also identified as independent contributing factors in multivariate models. Kaplan-Meier survival curves associated HO-1 expression with a poor prognosis in metastasis-free (P=0.047) and cause-specific survival (P=0.017), but not with urinary tract recurrence (P=0.231). Furthermore, HO-1 expression was identified by multivariate analysis to be a significant predictor for cause-specific survival (hazard ratio, 4.08; 95% confidence interval, 1.06-15.66; P=0.004). HO-1 has an important role in the malignant aggressiveness of NMIBC and its expression is associated with cause-specific survival. HO-1-associated activities are regulated by cancer cell proliferation, lymphangiogenesis and COX-2. The results suggest that HO-1 may be a potential therapeutic target and a useful predictive prognostic factor in patients with NMIBC.
Collapse
Affiliation(s)
- Tomohiro Matsuo
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yasuyoshi Miyata
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Kensuke Mitsunari
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Takuji Yasuda
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Kojiro Ohba
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Hideki Sakai
- Department of Urology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| |
Collapse
|
41
|
Halin Bergström S, Nilsson M, Adamo H, Thysell E, Jernberg E, Stattin P, Widmark A, Wikström P, Bergh A. Extratumoral Heme Oxygenase-1 (HO-1) Expressing Macrophages Likely Promote Primary and Metastatic Prostate Tumor Growth. PLoS One 2016; 11:e0157280. [PMID: 27280718 PMCID: PMC4900522 DOI: 10.1371/journal.pone.0157280] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 05/26/2016] [Indexed: 01/06/2023] Open
Abstract
Aggressive tumors induce tumor-supporting changes in the benign parts of the prostate. One factor that has increased expression outside prostate tumors is hemoxygenase-1 (HO-1). To investigate HO-1 expression in more detail, we analyzed samples of tumor tissue and peritumoral normal prostate tissue from rats carrying cancers with different metastatic capacity, and human prostate cancer tissue samples from primary tumors and bone metastases. In rat prostate tumor samples, immunohistochemistry and quantitative RT-PCR showed that the main site of HO-1 synthesis was HO-1+ macrophages that accumulated in the tumor-bearing organ, and at the tumor-invasive front. Small metastatic tumors were considerably more effective in attracting HO-1+ macrophages than larger non-metastatic ones. In clinical samples, accumulation of HO-1+ macrophages was seen at the tumor invasive front, almost exclusively in high-grade tumors, and it correlated with the presence of bone metastases. HO-1+ macrophages, located at the tumor invasive front, were more abundant in bone metastases than in primary tumors. HO-1 expression in bone metastases was variable, and positively correlated with the expression of macrophage markers but negatively correlated with androgen receptor expression, suggesting that elevated HO-1 could be a marker for a subgroup of bone metastases. Together with another recent observation showing that selective knockout of HO-1 in macrophages reduced prostate tumor growth and metastatic capacity in animals, the results of this study suggest that extratumoral HO-1+ macrophages may have an important role in prostate cancer.
Collapse
Affiliation(s)
- Sofia Halin Bergström
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
- * E-mail:
| | - Maria Nilsson
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Hanibal Adamo
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Elin Thysell
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Emma Jernberg
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Pär Stattin
- Department of Surgical and Perioperative Sciences, Urology, Umeå University, Umeå, Sweden
| | - Anders Widmark
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Pernilla Wikström
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Anders Bergh
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| |
Collapse
|
42
|
Kongpetch S, Puapairoj A, Ong CK, Senggunprai L, Prawan A, Kukongviriyapan U, Chan-On W, Siew EY, Khuntikeo N, Teh BT, Kukongviriyapan V. Haem oxygenase 1 expression is associated with prognosis in cholangiocarcinoma patients and with drug sensitivity in xenografted mice. Cell Prolif 2016; 49:90-101. [PMID: 26726846 DOI: 10.1111/cpr.12228] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 08/21/2015] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Haem oxygenase-1 (HO-1) plays important roles in cytoprotection and tumour growth. Cholangiocarcinoma (CCA) is a deadly malignancy with very poor prognosis. The role of HO-1 in tumour progression in CCA up to now has been relatively unexplored, thus, its possible therapeutic implications in CCA have been investigated here. MATERIALS AND METHODS HO-1 expression in tumour tissues from 50 CCA patients was determined by immunohistochemical analysis and its association with survival time was evaluated using the Kaplan-Meier method. Its role in CCA cells in vitro was evaluated by transwell and wound healing assays and suppression of HO-1 expression by siRNA. Effects of HO-1 inhibition on gemicitabine (GEM)-mediated tumour suppression was evaluated in nude mice xenografted with CCA cells. RESULTS HO-1 expression was inversely associated with median overall survival time. Hazard ratio of patients with high HO-1 expression was 2.42 (95% CI: 1.16-5.08) with reference to low expression and HO-1 knock-down expression inhibited transwell cell migration. Suppression of HO-1 by Zn-protoporphyrin (ZnPP) enhanced cytotoxicity to GEM in CCA cells, validated in CCA xenografts. Treatment with GEM and ZnPP almost completely arrested tumour growth, whereas treatment with only a single reagent, retarded it. Tumour inhibition was associated with reduction in expression of Ki-67 and microvascular density, and enhanced p53 and p21 immunohistochemical staining. CONCLUSION High HO-1 expression was associated with poor prognosis of CCA. Synergistic role of HO-1 inhibition in chemotherapy of CCA is a promising insight for treatment of this tumour and warrants further investigation.
Collapse
Affiliation(s)
- S Kongpetch
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand
| | - A Puapairoj
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - C K Ong
- Laboratory of Cancer Epigenome, National Cancer Centre of Singapore, Singapore.,Division of Cancer and Stem Cell Biology, Duke-National University of Singapore Graduate Medical School, Singapore
| | - L Senggunprai
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand
| | - A Prawan
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand
| | - U Kukongviriyapan
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - W Chan-On
- Laboratory of Cancer Epigenome, National Cancer Centre of Singapore, Singapore
| | - E Y Siew
- Laboratory of Cancer Epigenome, National Cancer Centre of Singapore, Singapore.,Division of Cancer and Stem Cell Biology, Duke-National University of Singapore Graduate Medical School, Singapore
| | - N Khuntikeo
- Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand.,Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - B T Teh
- Laboratory of Cancer Epigenome, National Cancer Centre of Singapore, Singapore.,Division of Cancer and Stem Cell Biology, Duke-National University of Singapore Graduate Medical School, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - V Kukongviriyapan
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
43
|
The Nrf2/HO-1 Axis in Cancer Cell Growth and Chemoresistance. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:1958174. [PMID: 26697129 PMCID: PMC4677237 DOI: 10.1155/2016/1958174] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 08/13/2015] [Accepted: 08/18/2015] [Indexed: 12/20/2022]
Abstract
The transcription factor, nuclear factor erythroid 2 p45-related factor 2 (Nrf2), acts as a sensor of oxidative or electrophilic stresses and plays a pivotal role in redox homeostasis. Oxidative or electrophilic agents cause a conformational change in the Nrf2 inhibitory protein Keap1 inducing the nuclear translocation of the transcription factor which, through its binding to the antioxidant/electrophilic response element (ARE/EpRE), regulates the expression of antioxidant and detoxifying genes such as heme oxygenase 1 (HO-1). Nrf2 and HO-1 are frequently upregulated in different types of tumours and correlate with tumour progression, aggressiveness, resistance to therapy, and poor prognosis. This review focuses on the Nrf2/HO-1 stress response mechanism as a promising target for anticancer treatment which is able to overcome resistance to therapies.
Collapse
|
44
|
Loboda A, Jozkowicz A, Dulak J. HO-1/CO system in tumor growth, angiogenesis and metabolism - Targeting HO-1 as an anti-tumor therapy. Vascul Pharmacol 2015; 74:11-22. [PMID: 26392237 DOI: 10.1016/j.vph.2015.09.004] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 09/02/2015] [Accepted: 09/16/2015] [Indexed: 02/08/2023]
Abstract
Heme oxygenase-1 (HO-1, hmox-1) catalyzes the rate-limiting step in the heme degradation processes. Out of three by-products of HO-1 activity, biliverdin, iron ions and carbon monoxide (CO), the latter was mostly shown to mediate many beneficial HO-1 effects, including protection against oxidative injury, regulation of apoptosis, modulation of inflammation as well as contribution to angiogenesis. Mounting evidence suggests that HO-1/CO systemmay be of special benefit in protection inmany pathological conditions, like atherosclerosis or myocardial infarction. By contrast, the augmented expression of HO-1 in tumor tissues may have detrimental effect as HO-1 accelerates the formation of tumor neovasculature and provides the selective advantage for tumor cells to overcome the increased oxidative stress during tumorigenesis and during treatment. The inhibition of HO-1 has been proposed as an anti-cancer therapy, however, because of non-specific effects of known HO-1 inhibitors, the discovery of ideal drug lowering HO-1 expression/activity is still an open question. Importantly, in several types of cancer HO-1/CO system exerts opposite activities, making the possible treatment more complicated. All together indicates the complex role for HO-1/CO in various in vitro and in vivo conditions.
Collapse
Affiliation(s)
- Agnieszka Loboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland; Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland.
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland; Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland.
| |
Collapse
|
45
|
Labanca E, De Luca P, Gueron G, Paez A, Moiola CP, Massillo C, Porretti J, Giudice J, Zalazar F, Navone N, Vazquez E, De Siervi A. Association of HO-1 and BRCA1 Is Critical for the Maintenance of Cellular Homeostasis in Prostate Cancer. Mol Cancer Res 2015; 13:1455-64. [PMID: 26227317 DOI: 10.1158/1541-7786.mcr-15-0150-t] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 07/22/2015] [Indexed: 11/16/2022]
Abstract
UNLABELLED Prostate cancer is the second leading cause of cancer-related death in men worldwide. Many factors that participate in the development of prostate cancer promote imbalance in the redox state of the cell. Accumulation of reactive oxygen species causes injury to cell structures, ultimately leading to cancer development. The antioxidant enzyme heme oxygenase 1 (HMOX1/HO-1) is responsible for the maintenance of the cellular homeostasis, playing a critical role in the oxidative stress and the regulation of prostate cancer development and progression. In the present study, the transcriptional regulation of HO-1 was investigated in prostate cancer. Interestingly, the tumor suppressor BRCA1 binds to the HO-1 promoter and modulates HO-1, inducing its protein levels through both the increment of its promoter activity and the induction of its transcriptional activation. In addition, in vitro and in vivo analyses show that BRCA1 also controls HO-1-negative targets: MMP9, uPA, and Cyclin D1. HO-1 transcriptional regulation is also modulated by oxidative and genotoxic agents. Induction of DNA damage by mitoxantrone and etoposide repressed HO-1 transcription, whereas hydrogen peroxide and doxorubicin induced its expression. Xenograft studies showed that HO-1 regulation by doxorubicin also occurs in vivo. Immunofluorescence analysis revealed that BRCA1 overexpression and/or doxorubicin exposure induced the cytoplasmic retention of HO-1. Finally, the transcription factor NRF2 cooperates with BRCA1 protein to activate HO-1 promoter activity. In summary, these results show that the activation of BRCA1-NRF2/HO-1 axis defines a new mechanism for the maintenance of the cellular homeostasis in prostate cancer. IMPLICATIONS Oxidative and genotoxic stress converge on HO-1 transcriptional activity through the combined actions of BRCA1 and NRF2.
Collapse
Affiliation(s)
- Estefanía Labanca
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biologia y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina. Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), IQUIBICEN-CONICET, Buenos Aires, Argentina
| | - Paola De Luca
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biologia y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina
| | - Geraldine Gueron
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), IQUIBICEN-CONICET, Buenos Aires, Argentina
| | - Alejandra Paez
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), IQUIBICEN-CONICET, Buenos Aires, Argentina
| | - Cristian P Moiola
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biologia y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina
| | - Cintia Massillo
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biologia y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina
| | - Juliana Porretti
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biologia y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina
| | - Jimena Giudice
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Florencia Zalazar
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biologia y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina
| | - Nora Navone
- Department of Genitourinary Medical Oncology, The University of Texas, M. D. Anderson Cancer Center, Houston, Texas
| | - Elba Vazquez
- Laboratorio de Inflamación y Cáncer, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales (FCEN), Universidad de Buenos Aires (UBA), IQUIBICEN-CONICET, Buenos Aires, Argentina
| | - Adriana De Siervi
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos, Instituto de Biologia y Medicina Experimental (IBYME)-CONICET, Buenos Aires, Argentina.
| |
Collapse
|
46
|
Gibbs PEM, Miralem T, Maines MD. Biliverdin reductase: a target for cancer therapy? Front Pharmacol 2015; 6:119. [PMID: 26089799 PMCID: PMC4452799 DOI: 10.3389/fphar.2015.00119] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 05/20/2015] [Indexed: 12/30/2022] Open
Abstract
Biliverdin reductase (BVR) is a multifunctional protein that is the primary source of the potent antioxidant, bilirubin. BVR regulates activities/functions in the insulin/IGF-1/IRK/PI3K/MAPK pathways. Activation of certain kinases in these pathways is/are hallmark(s) of cancerous cells. The protein is a scaffold/bridge and intracellular transporter of kinases that regulate growth and proliferation of cells, including PKCs, ERK and Akt, and their targets including NF-κB, Elk1, HO-1, and iNOS. The scaffold and transport functions enable activated BVR to relocate from the cytosol to the nucleus or to the plasma membrane, depending on the activating stimulus. This enables the reductase to function in diverse signaling pathways. And, its expression at the transcript and protein levels are increased in human tumors and the infiltrating T-cells, monocytes and circulating lymphocytes, as well as the circulating and infiltrating macrophages. These functions suggest that the cytoprotective role of BVR may be permissive for cancer/tumor growth. In this review, we summarize the recent developments that define the pro-growth activities of BVR, particularly with respect to its input into the MAPK signaling pathway and present evidence that BVR-based peptides inhibit activation of protein kinases, including MEK, PKCδ, and ERK as well as downstream targets including Elk1 and iNOS, and thus offers a credible novel approach to reduce cancer cell proliferation.
Collapse
Affiliation(s)
- Peter E M Gibbs
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry , Rochester, NY, USA
| | - Tihomir Miralem
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry , Rochester, NY, USA
| | - Mahin D Maines
- Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry , Rochester, NY, USA
| |
Collapse
|
47
|
Huang SM, Lin C, Lin HY, Chiu CM, Fang CW, Liao KF, Chen DR, Yeh WL. Brain-derived neurotrophic factor regulates cell motility in human colon cancer. Endocr Relat Cancer 2015; 22:455-64. [PMID: 25876647 DOI: 10.1530/erc-15-0007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/14/2015] [Indexed: 12/13/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a potent neurotrophic factor that has been shown to affect cancer cell metastasis and migration. In the present study, we investigated the mechanisms of BDNF-induced cell migration in colon cancer cells. The migratory activities of two colon cancer cell lines, HCT116 and SW480, were found to be increased in the presence of human BDNF. Heme oxygenase-1 (HO)-1 is known to be involved in the development and progression of tumors. However, the molecular mechanisms that underlie HO-1 in the regulation of colon cancer cell migration remain unclear. Expression of HO-1 protein and mRNA increased in response to BDNF stimulation. The BDNF-induced increase in cell migration was antagonized by a HO-1 inhibitor and HO-1 siRNA. Furthermore, the expression of vascular endothelial growth factor (VEGF) also increased in response to BDNF stimulation, as did VEGF mRNA expression and transcriptional activity. The increase in BDNF-induced cancer cell migration was antagonized by a VEGF-neutralizing antibody. Moreover, transfection with HO-1 siRNA effectively reduced the increased VEGF expression induced by BDNF. The BDNF-induced cell migration was regulated by the ERK, p38, and Akt signaling pathways. Furthermore, BDNF-increased HO-1 and VEGF promoter transcriptional activity were inhibited by ERK, p38, and AKT pharmacological inhibitors and dominant-negative mutants in colon cancer cells. These results indicate that BDNF increases the migration of colon cancer cells by regulating VEGF/HO-1 activation through the ERK, p38, and PI3K/Akt signaling pathways. The results of this study may provide a relevant contribution to our understanding of the molecular mechanisms by which BDNF promotes colon cancer cell motility.
Collapse
Affiliation(s)
- Ssu-Ming Huang
- Department of Community MedicinePreventive Medicine CenterDivision of Colon and Rectal SurgeryDepartment of Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanSchool of MedicineTzu Chi University, Hualien, TaiwanDepartment of PhysiologySchool of MedicineGraduate Institute of Neural and Cognitive SciencesChina Medical University, Taichung, TaiwanDepartment of Internal MedicineTaichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanDepartment of Chinese MedicineGraduate Institute of Integrated Medicine, China Medical University, Taichung, TaiwanComprehensive Breast Cancer CenterDepartment of Cell and Tissue EngineeringChanghua Christian Hospital, Nanxiao St., Changhua City, Changhua County 500, Taiwan Department of Community MedicinePreventive Medicine CenterDivision of Colon and Rectal SurgeryDepartment of Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanSchool of MedicineTzu Chi University, Hualien, TaiwanDepartment of PhysiologySchool of MedicineGraduate Institute of Neural and Cognitive SciencesChina Medical University, Taichung, TaiwanDepartment of Internal MedicineTaichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanDepartment of Chinese MedicineGraduate Institute of Integrated Medicine, China Medical University, Taichung, TaiwanComprehensive Breast Cancer CenterDepartment of Cell and Tissue EngineeringChanghua Christian Hospital, Nanxiao St., Changhua City, Changhua County 500, Taiwan Department of Community MedicinePreventive Medicine CenterDivision of Colon and Rectal SurgeryDepartment of Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanSchool of MedicineTzu Chi University, Hualien, TaiwanDepartment of PhysiologySchool of MedicineGraduate Institute of Neural and Cognitive SciencesChina Medical University, Taichung, TaiwanDepartment of Internal MedicineTaichung Tzu Chi Hospital, Buddhist Tzu Ch
| | - Chingju Lin
- Department of Community MedicinePreventive Medicine CenterDivision of Colon and Rectal SurgeryDepartment of Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanSchool of MedicineTzu Chi University, Hualien, TaiwanDepartment of PhysiologySchool of MedicineGraduate Institute of Neural and Cognitive SciencesChina Medical University, Taichung, TaiwanDepartment of Internal MedicineTaichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanDepartment of Chinese MedicineGraduate Institute of Integrated Medicine, China Medical University, Taichung, TaiwanComprehensive Breast Cancer CenterDepartment of Cell and Tissue EngineeringChanghua Christian Hospital, Nanxiao St., Changhua City, Changhua County 500, Taiwan
| | - Hsiao-Yun Lin
- Department of Community MedicinePreventive Medicine CenterDivision of Colon and Rectal SurgeryDepartment of Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanSchool of MedicineTzu Chi University, Hualien, TaiwanDepartment of PhysiologySchool of MedicineGraduate Institute of Neural and Cognitive SciencesChina Medical University, Taichung, TaiwanDepartment of Internal MedicineTaichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanDepartment of Chinese MedicineGraduate Institute of Integrated Medicine, China Medical University, Taichung, TaiwanComprehensive Breast Cancer CenterDepartment of Cell and Tissue EngineeringChanghua Christian Hospital, Nanxiao St., Changhua City, Changhua County 500, Taiwan
| | - Chien-Ming Chiu
- Department of Community MedicinePreventive Medicine CenterDivision of Colon and Rectal SurgeryDepartment of Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanSchool of MedicineTzu Chi University, Hualien, TaiwanDepartment of PhysiologySchool of MedicineGraduate Institute of Neural and Cognitive SciencesChina Medical University, Taichung, TaiwanDepartment of Internal MedicineTaichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanDepartment of Chinese MedicineGraduate Institute of Integrated Medicine, China Medical University, Taichung, TaiwanComprehensive Breast Cancer CenterDepartment of Cell and Tissue EngineeringChanghua Christian Hospital, Nanxiao St., Changhua City, Changhua County 500, Taiwan
| | - Chia-Wei Fang
- Department of Community MedicinePreventive Medicine CenterDivision of Colon and Rectal SurgeryDepartment of Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanSchool of MedicineTzu Chi University, Hualien, TaiwanDepartment of PhysiologySchool of MedicineGraduate Institute of Neural and Cognitive SciencesChina Medical University, Taichung, TaiwanDepartment of Internal MedicineTaichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanDepartment of Chinese MedicineGraduate Institute of Integrated Medicine, China Medical University, Taichung, TaiwanComprehensive Breast Cancer CenterDepartment of Cell and Tissue EngineeringChanghua Christian Hospital, Nanxiao St., Changhua City, Changhua County 500, Taiwan
| | - Kuan-Fu Liao
- Department of Community MedicinePreventive Medicine CenterDivision of Colon and Rectal SurgeryDepartment of Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanSchool of MedicineTzu Chi University, Hualien, TaiwanDepartment of PhysiologySchool of MedicineGraduate Institute of Neural and Cognitive SciencesChina Medical University, Taichung, TaiwanDepartment of Internal MedicineTaichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanDepartment of Chinese MedicineGraduate Institute of Integrated Medicine, China Medical University, Taichung, TaiwanComprehensive Breast Cancer CenterDepartment of Cell and Tissue EngineeringChanghua Christian Hospital, Nanxiao St., Changhua City, Changhua County 500, Taiwan Department of Community MedicinePreventive Medicine CenterDivision of Colon and Rectal SurgeryDepartment of Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanSchool of MedicineTzu Chi University, Hualien, TaiwanDepartment of PhysiologySchool of MedicineGraduate Institute of Neural and Cognitive SciencesChina Medical University, Taichung, TaiwanDepartment of Internal MedicineTaichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanDepartment of Chinese MedicineGraduate Institute of Integrated Medicine, China Medical University, Taichung, TaiwanComprehensive Breast Cancer CenterDepartment of Cell and Tissue EngineeringChanghua Christian Hospital, Nanxiao St., Changhua City, Changhua County 500, Taiwan Department of Community MedicinePreventive Medicine CenterDivision of Colon and Rectal SurgeryDepartment of Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanSchool of MedicineTzu Chi University, Hualien, TaiwanDepartment of PhysiologySchool of MedicineGraduate Institute of Neural and Cognitive SciencesChina Medical University, Taichung, TaiwanDepartment of Internal MedicineTaichung Tzu Chi Hospital, Buddhist Tzu Ch
| | - Dar-Ren Chen
- Department of Community MedicinePreventive Medicine CenterDivision of Colon and Rectal SurgeryDepartment of Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanSchool of MedicineTzu Chi University, Hualien, TaiwanDepartment of PhysiologySchool of MedicineGraduate Institute of Neural and Cognitive SciencesChina Medical University, Taichung, TaiwanDepartment of Internal MedicineTaichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanDepartment of Chinese MedicineGraduate Institute of Integrated Medicine, China Medical University, Taichung, TaiwanComprehensive Breast Cancer CenterDepartment of Cell and Tissue EngineeringChanghua Christian Hospital, Nanxiao St., Changhua City, Changhua County 500, Taiwan
| | - Wei-Lan Yeh
- Department of Community MedicinePreventive Medicine CenterDivision of Colon and Rectal SurgeryDepartment of Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanSchool of MedicineTzu Chi University, Hualien, TaiwanDepartment of PhysiologySchool of MedicineGraduate Institute of Neural and Cognitive SciencesChina Medical University, Taichung, TaiwanDepartment of Internal MedicineTaichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, TaiwanDepartment of Chinese MedicineGraduate Institute of Integrated Medicine, China Medical University, Taichung, TaiwanComprehensive Breast Cancer CenterDepartment of Cell and Tissue EngineeringChanghua Christian Hospital, Nanxiao St., Changhua City, Changhua County 500, Taiwan
| |
Collapse
|
48
|
Gueron G, Giudice J, Valacco P, Paez A, Elguero B, Toscani M, Jaworski F, Leskow FC, Cotignola J, Marti M, Binaghi M, Navone N, Vazquez E. Heme-oxygenase-1 implications in cell morphology and the adhesive behavior of prostate cancer cells. Oncotarget 2015; 5:4087-102. [PMID: 24961479 PMCID: PMC4147308 DOI: 10.18632/oncotarget.1826] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer death in men. Although previous studies in PCa have focused on cell adherens junctions (AJs), key players in metastasis, they have left the molecular mechanisms unexplored. Inflammation and the involvement of reactive oxygen species (ROS) are critical in the regulation of cell adhesion and the integrity of the epithelium. Heme oxygenase-1 (HO-1) counteracts oxidative and inflammatory damage. Here, we investigated whether HO-1 is implicated in the adhesive and morphological properties of tumor cells. Genes differentially regulated by HO-1 were enriched for cell motility and adhesion biological processes. HO-1 induction, increased E-cadherin and β-catenin levels. Immunofluorescence analyses showed a striking remodeling of E-cadherin/β-catenin based AJs under HO-1 modulation. Interestingly, the enhanced levels of E-cadherin and β-catenin coincided with a markedly change in cell morphology. To further our analysis we sought to identify HO-1 binding proteins that might participate in the regulation of cell morphology. A proteomics approach identified Muskelin, as a novel HO-1 partner, strongly implicated in cell morphology regulation. These results define a novel role for HO-1 in modulating the architecture of cell-cell interactions, favoring a less aggressive phenotype and further supporting its anti-tumoral function in PCa.
Collapse
|
49
|
Heme oxygenase-1 expression is associated with tumor aggressiveness and outcomes in patients with bladder cancer: a correlation with smoking intensity. Transl Res 2014; 164:468-76. [PMID: 25063314 DOI: 10.1016/j.trsl.2014.06.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 05/28/2014] [Accepted: 06/26/2014] [Indexed: 12/21/2022]
Abstract
Heme oxygenase (HO)-1 is upregulated in malignancies and, in turn, regulates other cancer-related factors. Although HO-1 expression has been associated with cigarette smoking under various pathologic conditions, little is known about their association in patients with bladder cancer. HO-1 expression was assessed in 215 formalin-fixed bladder cancer specimens by immunohistochemistry. Microvessel density, lymph vessel density (LVD), proliferation index (PI), and expression of the vascular endothelial growth factor (VEGF)-A, -C, and -D, cyclooxygenase (COX)-2, matrix metalloproteinase (MMP)-2, and MMP-9 were investigated by similar methods. Multivariate analyses were performed to evaluate the pathologic role and predictive value of HO-1 expression. Our results demonstrated that HO-1 expression was positively associated with T stage, lymph node metastasis, and grade. HO-1 expression was also positively correlated with PI, LVD, and expression levels of VEGF-D, COX-2, MMP-2, and MMP-9 (P < 0.001). In addition, multivariate analyses showed that HO-1 expression positively correlated with smoking intensity. Positive HO-1 expression was a significant predictor of subsequent metastasis (P = 0.008) and poor cause-specific survival (P < 0.001). Similarly, multivariate analyses showed that HO-1 expression was a predictor of cause-specific survival (hazard ratio = 3.13, P = 0.013). In conclusion, pathologic changes of HO-1-related factors were dependent on smoking intensity. Smoking upregulated HO-1 expression, and HO-1 was associated with malignant behavior of bladder cancer. Cancer cell proliferation, lymphangiogenesis, and expression levels of VEGF-D, COX-2, and MMP-2 played important roles in these HO-1-related effects. The clinical correlations of HO-1 were regulated by a complex mechanism that depended on smoking intensity.
Collapse
|
50
|
Sterle HA, Valli E, Cayrol F, Paulazo MA, Martinel Lamas DJ, Diaz Flaqué MC, Klecha AJ, Colombo L, Medina VA, Cremaschi GA, Barreiro Arcos ML. Thyroid status modulates T lymphoma growth via cell cycle regulatory proteins and angiogenesis. J Endocrinol 2014; 222:243-55. [PMID: 24928937 DOI: 10.1530/joe-14-0159] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We have shown in vitro that thyroid hormones (THs) regulate the balance between proliferation and apoptosis of T lymphoma cells. The effects of THs on tumor development have been studied, but the results are still controversial. Herein, we show the modulatory action of thyroid status on the in vivo growth of T lymphoma cells. For this purpose, euthyroid, hypothyroid, and hyperthyroid mice received inoculations of EL4 cells to allow the development of solid tumors. Tumors in the hyperthyroid animals exhibited a higher growth rate, as evidenced by the early appearance of palpable solid tumors and the increased tumor volume. These results are consistent with the rate of cell division determined by staining tumor cells with carboxyfluorescein succinimidyl ester. Additionally, hyperthyroid mice exhibited reduced survival. Hypothyroid mice did not differ significantly from the euthyroid controls with respect to these parameters. Additionally, only tumors from hyperthyroid animals had increased expression levels of proliferating cell nuclear antigen and active caspase 3. Differential expression of cell cycle regulatory proteins was also observed. The levels of cyclins D1 and D3 were augmented in the tumors of the hyperthyroid animals, whereas the cell cycle inhibitors p16/INK4A (CDKN2A) and p27/Kip1 (CDKN1B) and the tumor suppressor p53 (TRP53) were increased in hypothyroid mice. Intratumoral and peritumoral vasculogenesis was increased only in hyperthyroid mice. Therefore, we propose that the thyroid status modulates the in vivo growth of EL4 T lymphoma through the regulation of cyclin, cyclin-dependent kinase inhibitor, and tumor suppressor gene expression, as well as the stimulation of angiogenesis.
Collapse
Affiliation(s)
- H A Sterle
- Instituto de Investigaciones Biomédicas (BIOMED)Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina (UCA), Av. A. Moreau de Justo 1600, 3er piso, 1107AFF Buenos Aires, ArgentinaCentro de Estudios Farmacológicos y Botánicos (CEFYBO)CONICET, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaLaboratorio de RadioisótoposFacultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaArea de InvestigaciónInstituto de Oncología 'Angel H. Roffo', Universidad de Buenos Aires (UBA), CONICET, Buenos Aires, ArgentinaDepartamento de Química BiológicaFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - E Valli
- Instituto de Investigaciones Biomédicas (BIOMED)Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina (UCA), Av. A. Moreau de Justo 1600, 3er piso, 1107AFF Buenos Aires, ArgentinaCentro de Estudios Farmacológicos y Botánicos (CEFYBO)CONICET, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaLaboratorio de RadioisótoposFacultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaArea de InvestigaciónInstituto de Oncología 'Angel H. Roffo', Universidad de Buenos Aires (UBA), CONICET, Buenos Aires, ArgentinaDepartamento de Química BiológicaFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - F Cayrol
- Instituto de Investigaciones Biomédicas (BIOMED)Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina (UCA), Av. A. Moreau de Justo 1600, 3er piso, 1107AFF Buenos Aires, ArgentinaCentro de Estudios Farmacológicos y Botánicos (CEFYBO)CONICET, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaLaboratorio de RadioisótoposFacultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaArea de InvestigaciónInstituto de Oncología 'Angel H. Roffo', Universidad de Buenos Aires (UBA), CONICET, Buenos Aires, ArgentinaDepartamento de Química BiológicaFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - M A Paulazo
- Instituto de Investigaciones Biomédicas (BIOMED)Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina (UCA), Av. A. Moreau de Justo 1600, 3er piso, 1107AFF Buenos Aires, ArgentinaCentro de Estudios Farmacológicos y Botánicos (CEFYBO)CONICET, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaLaboratorio de RadioisótoposFacultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaArea de InvestigaciónInstituto de Oncología 'Angel H. Roffo', Universidad de Buenos Aires (UBA), CONICET, Buenos Aires, ArgentinaDepartamento de Química BiológicaFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - D J Martinel Lamas
- Instituto de Investigaciones Biomédicas (BIOMED)Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina (UCA), Av. A. Moreau de Justo 1600, 3er piso, 1107AFF Buenos Aires, ArgentinaCentro de Estudios Farmacológicos y Botánicos (CEFYBO)CONICET, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaLaboratorio de RadioisótoposFacultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaArea de InvestigaciónInstituto de Oncología 'Angel H. Roffo', Universidad de Buenos Aires (UBA), CONICET, Buenos Aires, ArgentinaDepartamento de Química BiológicaFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - M C Diaz Flaqué
- Instituto de Investigaciones Biomédicas (BIOMED)Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina (UCA), Av. A. Moreau de Justo 1600, 3er piso, 1107AFF Buenos Aires, ArgentinaCentro de Estudios Farmacológicos y Botánicos (CEFYBO)CONICET, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaLaboratorio de RadioisótoposFacultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaArea de InvestigaciónInstituto de Oncología 'Angel H. Roffo', Universidad de Buenos Aires (UBA), CONICET, Buenos Aires, ArgentinaDepartamento de Química BiológicaFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - A J Klecha
- Instituto de Investigaciones Biomédicas (BIOMED)Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina (UCA), Av. A. Moreau de Justo 1600, 3er piso, 1107AFF Buenos Aires, ArgentinaCentro de Estudios Farmacológicos y Botánicos (CEFYBO)CONICET, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaLaboratorio de RadioisótoposFacultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaArea de InvestigaciónInstituto de Oncología 'Angel H. Roffo', Universidad de Buenos Aires (UBA), CONICET, Buenos Aires, ArgentinaDepartamento de Química BiológicaFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - L Colombo
- Instituto de Investigaciones Biomédicas (BIOMED)Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina (UCA), Av. A. Moreau de Justo 1600, 3er piso, 1107AFF Buenos Aires, ArgentinaCentro de Estudios Farmacológicos y Botánicos (CEFYBO)CONICET, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaLaboratorio de RadioisótoposFacultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaArea de InvestigaciónInstituto de Oncología 'Angel H. Roffo', Universidad de Buenos Aires (UBA), CONICET, Buenos Aires, ArgentinaDepartamento de Química BiológicaFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - V A Medina
- Instituto de Investigaciones Biomédicas (BIOMED)Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina (UCA), Av. A. Moreau de Justo 1600, 3er piso, 1107AFF Buenos Aires, ArgentinaCentro de Estudios Farmacológicos y Botánicos (CEFYBO)CONICET, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaLaboratorio de RadioisótoposFacultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaArea de InvestigaciónInstituto de Oncología 'Angel H. Roffo', Universidad de Buenos Aires (UBA), CONICET, Buenos Aires, ArgentinaDepartamento de Química BiológicaFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - G A Cremaschi
- Instituto de Investigaciones Biomédicas (BIOMED)Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina (UCA), Av. A. Moreau de Justo 1600, 3er piso, 1107AFF Buenos Aires, ArgentinaCentro de Estudios Farmacológicos y Botánicos (CEFYBO)CONICET, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaLaboratorio de RadioisótoposFacultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaArea de InvestigaciónInstituto de Oncología 'Angel H. Roffo', Universidad de Buenos Aires (UBA), CONICET, Buenos Aires, ArgentinaDepartamento de Química BiológicaFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaInstituto de Investigaciones Biomédicas (BIOMED)Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina (UCA), Av. A. Moreau de Justo 1600, 3er piso, 1107AFF Buenos Aires, ArgentinaCentro de Estudios Farmacológicos y Botánicos (CEFYBO)CONICET, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaLaboratorio de RadioisótoposFacultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaArea de InvestigaciónInstituto de Oncología 'Angel H. Roffo', Universidad de Buenos Aires (UBA), CONICET, Buenos Aires, ArgentinaDepartamento de Química BiológicaFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - M L Barreiro Arcos
- Instituto de Investigaciones Biomédicas (BIOMED)Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina (UCA), Av. A. Moreau de Justo 1600, 3er piso, 1107AFF Buenos Aires, ArgentinaCentro de Estudios Farmacológicos y Botánicos (CEFYBO)CONICET, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaLaboratorio de RadioisótoposFacultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaArea de InvestigaciónInstituto de Oncología 'Angel H. Roffo', Universidad de Buenos Aires (UBA), CONICET, Buenos Aires, ArgentinaDepartamento de Química BiológicaFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaInstituto de Investigaciones Biomédicas (BIOMED)Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina (UCA), Av. A. Moreau de Justo 1600, 3er piso, 1107AFF Buenos Aires, ArgentinaCentro de Estudios Farmacológicos y Botánicos (CEFYBO)CONICET, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaLaboratorio de RadioisótoposFacultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, ArgentinaArea de InvestigaciónInstituto de Oncología 'Angel H. Roffo', Universidad de Buenos Aires (UBA), CONICET, Buenos Aires, ArgentinaDepartamento de Química BiológicaFacultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| |
Collapse
|