1
|
Pulous FE, Steurer B, Pun FW, Zhang M, Ren F, Zhavoronkov A. MAT2A inhibition combats metabolic and transcriptional reprogramming in cancer. Drug Discov Today 2024; 29:104189. [PMID: 39306235 DOI: 10.1016/j.drudis.2024.104189] [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: 07/02/2024] [Revised: 09/05/2024] [Accepted: 09/17/2024] [Indexed: 09/26/2024]
Abstract
Metabolic and transcriptional reprogramming are crucial hallmarks of carcinogenesis that present exploitable vulnerabilities for the development of targeted anticancer therapies. Through controlling the balance of the cellular methionine (MET) metabolite pool, MET adenosyl transferase 2 alpha (MAT2A) regulates crucial steps during metabolism and the epigenetic control of transcription. The aberrant function of MAT2A has been shown to drive malignant transformation through metabolic addiction, transcriptional rewiring, and immune modulation of the tumor microenvironment (TME). Moreover, MAT2A sustains the survival of 5'-methylthioadenosine phosphorylase (MTAP)-deficient tumors, conferring synthetic lethality to cancers with MTAP loss, a genetic alteration that occurs in ∼15% of all cancers. Thus, the pharmacological inhibition of MAT2A is emerging as a desirable therapeutic strategy to combat tumor growth. Here, we review the latest insights into MAT2A biology, focusing on its roles in both metabolic addiction and gene expression modulation in the TME, outline the current landscape of MAT2A inhibitors, and highlight the most recent clinical developments and opportunities for MAT2A inhibition as a novel anti-tumor therapy.
Collapse
Affiliation(s)
- Fadi E Pulous
- Insilico Medicine US Inc, 1000 Massachusetts Avenue, Suite 126, Cambridge, MA 02138, USA
| | - Barbara Steurer
- Insilico Medicine US Inc, 1000 Massachusetts Avenue, Suite 126, Cambridge, MA 02138, USA
| | - Frank W Pun
- Insilico Medicine Hong Kong Ltd, Unit 310, 3/F, Building 8W, Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - Man Zhang
- Insilico Medicine Shanghai Ltd, 9F, Chamtime Plaza Block C, Lane 2889, Jinke Road, Pudong New Area, China
| | - Feng Ren
- Insilico Medicine Shanghai Ltd, 9F, Chamtime Plaza Block C, Lane 2889, Jinke Road, Pudong New Area, China
| | - Alex Zhavoronkov
- Insilico Medicine US Inc, 1000 Massachusetts Avenue, Suite 126, Cambridge, MA 02138, USA; Insilico Medicine Hong Kong Ltd, Unit 310, 3/F, Building 8W, Hong Kong Science and Technology Park, Hong Kong SAR, China; Insilico Medicine AI Ltd, Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City, Abu Dhabi, UAE.
| |
Collapse
|
2
|
Ghareghomi S, Moosavi-Movahedi F, Saso L, Habibi-Rezaei M, Khatibi A, Hong J, Moosavi-Movahedi AA. Modulation of Nrf2/HO-1 by Natural Compounds in Lung Cancer. Antioxidants (Basel) 2023; 12:antiox12030735. [PMID: 36978983 PMCID: PMC10044870 DOI: 10.3390/antiox12030735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023] Open
Abstract
Oxidative stresses (OSs) are considered a pivotal factor in creating various pathophysiological conditions. Cells have been able to move forward by modulating numerous signaling pathways to moderate the defects of these stresses during their evolution. The company of Kelch-like ECH-associated protein 1 (Keap1) as a molecular sensing element of the oxidative and electrophilic stress and nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2) as a master transcriptional regulator of the antioxidant response makes a master cytoprotective antioxidant pathway known as the Keap1/Nrf2 pathway. This pathway is considered a dual-edged sword with beneficial features for both normal and cancer cells by regulating the gene expression of the array of endogenous antioxidant enzymes. Heme oxygenase-1 (HO-1), a critical enzyme in toxic heme removal, is one of the clear state indicators for the duality of this pathway. Therefore, Nrf2/HO-1 axis targeting is known as a novel strategy for cancer treatment. In this review, the molecular mechanism of action of natural antioxidants on lung cancer cells has been investigated by relying on the Nrf2/HO-1 axis.
Collapse
Affiliation(s)
- Somayyeh Ghareghomi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417466191, Iran; (S.G.); (F.M.-M.)
| | - Faezeh Moosavi-Movahedi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417466191, Iran; (S.G.); (F.M.-M.)
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence: (L.S.); (M.H.-R.); (A.A.M.-M.); Tel.: +39-06-4991-2481 (L.S.); +98-21-6111-3214 (M.H.-R.); +98-21-6640-3957 (A.A.M.-M.); Fax: +39-06-4991-2481 (L.S.); +98-21-6697-1941 (M.H.-R.); +98-21-6640-4680(A.A.M.-M.)
| | - Mehran Habibi-Rezaei
- School of Biology, College of Science, University of Tehran, Tehran 1417466191, Iran
- Center of Excellence in NanoBiomedicine, University of Tehran, Tehran 1417466191, Iran
- Correspondence: (L.S.); (M.H.-R.); (A.A.M.-M.); Tel.: +39-06-4991-2481 (L.S.); +98-21-6111-3214 (M.H.-R.); +98-21-6640-3957 (A.A.M.-M.); Fax: +39-06-4991-2481 (L.S.); +98-21-6697-1941 (M.H.-R.); +98-21-6640-4680(A.A.M.-M.)
| | - Ali Khatibi
- Department of Biotechnology, Faculty of Biological Sciences, Alzahra University, Tehran 1993893973, Iran;
| | - Jun Hong
- School of Life Sciences, Henan University, Kaifeng 475000, China;
| | - Ali A. Moosavi-Movahedi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417466191, Iran; (S.G.); (F.M.-M.)
- UNESCO Chair on Interdisciplinary Research in Diabetes, University of Tehran, Tehran 1417466191, Iran
- Correspondence: (L.S.); (M.H.-R.); (A.A.M.-M.); Tel.: +39-06-4991-2481 (L.S.); +98-21-6111-3214 (M.H.-R.); +98-21-6640-3957 (A.A.M.-M.); Fax: +39-06-4991-2481 (L.S.); +98-21-6697-1941 (M.H.-R.); +98-21-6640-4680(A.A.M.-M.)
| |
Collapse
|
3
|
The Role of HO-1 and Its Crosstalk with Oxidative Stress in Cancer Cell Survival. Cells 2021; 10:cells10092401. [PMID: 34572050 PMCID: PMC8471703 DOI: 10.3390/cells10092401] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 12/19/2022] Open
Abstract
Heme oxygenases (HOs) act on heme degradation to produce carbon monoxide (CO), free iron, ferritin, and biliverdin. Upregulation of cellular HO-1 levels is signature of oxidative stress for its downstream effects particularly under pro-oxidative status. Subcellular traffics of HO-1 to different organelles constitute a network of interactions compromising a variety of effectors such as pro-oxidants, ROS, mitochondrial enzymes, and nucleic transcription factors. Some of the compartmentalized HO-1 have been demonstrated as functioning in the progression of cancer. Emerging data show the multiple roles of HO-1 in tumorigenesis from pathogenesis to the progression to malignancy, metastasis, and even resistance to therapy. However, the role of HO-1 in tumorigenesis has not been systematically addressed. This review describes the crosstalk between HO-1 and oxidative stress, and following redox regulation in the tumorigenesis. HO-1-regulated signaling pathways are also summarized. This review aims to integrate basic information and current progress of HO-1 in cancer research in order to enhance the understandings and facilitate following studies.
Collapse
|
4
|
Miquelianin Inhibits Allergic Responses in Mice by Suppressing CD4 + T Cell Proliferation. Antioxidants (Basel) 2021; 10:antiox10071120. [PMID: 34356353 PMCID: PMC8301087 DOI: 10.3390/antiox10071120] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/05/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Allergic diseases, including atopic dermatitis (AD), induce type 2 helper T (Th2) cell-dominant immune responses. Miquelianin (quercetin 3-O-glucuronide, MQL) is an active compound in Rosae multiflorae fructus extract with anti-allergic properties. Here, we investigate the anti-allergic effects of MQL in an ovalbumin (OVA)-induced Th2-dominant mouse model and the associated mechanisms. Oral MQL suppressed cytokine and IL-2 production and proliferation of Th2 cells and upregulated heme oxygenase-1 (HO-1) in splenocytes. Ex vivo MQL suppressed Th1- and Th2-related immune responses by inhibiting CD4+ T cell proliferation, and upregulated HO-1 in CD4+ T cells by activating C-Raf-ERK1/2-Nrf2 pathway via induction of reactive oxygen species generation. In a trimellitic anhydride-induced AD-like mouse model, both topical and oral MQL ameliorated AD symptoms by suppressing Th2 immune responses. Our results suggest that MQL is a potential therapeutic agent for CD4+ T cell-mediated diseases, including allergic diseases.
Collapse
|
5
|
Panieri E, Saso L. Inhibition of the NRF2/KEAP1 Axis: A Promising Therapeutic Strategy to Alter Redox Balance of Cancer Cells. Antioxid Redox Signal 2021; 34:1428-1483. [PMID: 33403898 DOI: 10.1089/ars.2020.8146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: The nuclear factor erythroid 2-related factor 2/Kelch-like ECH-associated protein 1 (NRF2/KEAP1) pathway is a crucial and highly conserved defensive system that is required to maintain or restore the intracellular homeostasis in response to oxidative, electrophilic, and other types of stress conditions. The tight control of NRF2 function is maintained by a complex network of biological interactions between positive and negative regulators that ultimately ensure context-specific activation, culminating in the NRF2-driven transcription of cytoprotective genes. Recent Advances: Recent studies indicate that deregulated NRF2 activation is a frequent event in malignant tumors, wherein it is associated with metabolic reprogramming, increased antioxidant capacity, chemoresistance, and poor clinical outcome. On the other hand, the growing interest in the modulation of the cancer cells' redox balance identified NRF2 as an ideal therapeutic target. Critical Issues: For this reason, many efforts have been made to identify potent and selective NRF2 inhibitors that might be used as single agents or adjuvants of anticancer drugs with redox disrupting properties. Despite the lack of specific NRF2 inhibitors still represents a major clinical hurdle, the researchers have exploited alternative strategies to disrupt NRF2 signaling at different levels of its biological activation. Future Directions: Given its dualistic role in tumor initiation and progression, the identification of the appropriate biological context of NRF2 activation and the specific clinicopathological features of patients cohorts wherein its inactivation is expected to have clinical benefits, will represent a major goal in the field of cancer research. In this review, we will briefly describe the structure and function of the NRF2/ KEAP1 system and some of the most promising NRF2 inhibitors, with a particular emphasis on natural compounds and drug repurposing. Antioxid. Redox Signal. 34, 1428-1483.
Collapse
Affiliation(s)
- Emiliano Panieri
- Department of Physiology and Pharmacology "Vittorio Erspamer," University of Rome La Sapienza, Rome, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer," University of Rome La Sapienza, Rome, Italy
| |
Collapse
|
6
|
Kim EH, Kim SJ, Na HK, Han W, Kim NJ, Suh YG, Surh YJ. 15-Deoxy-Δ 12,14-prostaglandin J 2 Upregulates VEGF Expression via NRF2 and Heme Oxygenase-1 in Human Breast Cancer Cells. Cells 2021; 10:cells10030526. [PMID: 33801351 PMCID: PMC8002112 DOI: 10.3390/cells10030526] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
There is a plethora of evidence to support that inflammation is causally linked to carcinogenesis. Cyclooxygenase-2 (COX-2), a rate-limiting enzyme in the biosynthesis of prostaglandins, is inappropriately overexpressed in various cancers and hence recognized as one of the hallmarks of chronic inflammation-associated malignancies. However, the mechanistic role of COX-2 as a link between inflammation and cancer remains largely undefined. In this study, we found that 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2), one of the final products of COX-2, induced upregulation of vascular endothelial growth factor (VEGF) and capillary formation and migration through nuclear factor erythroid 2-related factor 2 (NRF2)-dependent heme oxygenase-1 (HO-1) induction in MCF-7 cells. Analysis of the publicly available TCGA data set showed that high mRNA levels of both COX-2 and NRF2 correlated with the poor clinical outcomes in breast cancer patients. Moreover, human tissue analysis showed that the levels of 15d-PGJ2 as well the expression of COX-2, NRF2, and HO-1 were found to be increased in human breast cancer tissues. In conclusion, the elevated levels of 15d-PGJ2 during inflammatory response activate VEGF expression through NRF2-driven induction of HO-1 in human breast cancer cells, proposing a novel mechanism underlying the oncogenic function of 15d-PGJ2.
Collapse
Affiliation(s)
- Eun-Hee Kim
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam 13488, Korea;
- Correspondence: (E.-H.K.); (Y.-J.S.); Tel.: +82-31-881-7179 (E.-H.K.); +82-2-880-7845 (Y.-J.S.)
| | - Su-Jung Kim
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, Korea;
| | - Hye-Kyung Na
- Department of Food Science and Biotechnology, College of Knowledge-Based Services Engineering, Sungshin Women’s University, Seoul 01133, Korea;
| | - Wonshik Han
- Cancer Research Institute, Seoul National University, Seoul 03080, Korea;
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Nam-Jung Kim
- College of Pharmacy, Kyung Hee University, Seoul 02447, Korea;
| | - Young-Ger Suh
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam 13488, Korea;
| | - Young-Joon Surh
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul 08826, Korea;
- Cancer Research Institute, Seoul National University, Seoul 03080, Korea;
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Korea
- Correspondence: (E.-H.K.); (Y.-J.S.); Tel.: +82-31-881-7179 (E.-H.K.); +82-2-880-7845 (Y.-J.S.)
| |
Collapse
|
7
|
Hahn D, Shin SH, Bae JS. Natural Antioxidant and Anti-Inflammatory Compounds in Foodstuff or Medicinal Herbs Inducing Heme Oxygenase-1 Expression. Antioxidants (Basel) 2020; 9:E1191. [PMID: 33260980 PMCID: PMC7761319 DOI: 10.3390/antiox9121191] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023] Open
Abstract
Heme oxygenase-1 (HO-1) is an inducible antioxidant enzyme that catalyzes heme group degradation. Decreased level of HO-1 is correlated with disease progression, and HO-1 induction suppresses development of metabolic and neurological disorders. Natural compounds with antioxidant activities have emerged as a rich source of HO-1 inducers with marginal toxicity. Here we discuss the therapeutic role of HO-1 in obesity, hypertension, atherosclerosis, Parkinson's disease and hepatic fibrosis, and present important signaling pathway components that lead to HO-1 expression. We provide an updated, comprehensive list of natural HO-1 inducers in foodstuff and medicinal herbs categorized by their chemical structures. Based on the continued research in HO-1 signaling pathways and rapid development of their natural inducers, HO-1 may serve as a preventive and therapeutic target for metabolic and neurological disorders.
Collapse
Affiliation(s)
- Dongyup Hahn
- School of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Korea;
- Department of Integrative Biology, Kyungpook National University, Daegu 41566, Korea
| | - Seung Ho Shin
- Department of Food and Nutrition, Institute of Agriculture and Life Science, Gyeongsang National University, Jinju 52828, Korea;
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, BK21 Plus KNU Multi-Omics based Creative Drug Research Team, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
8
|
Clerici S, Boletta A. Role of the KEAP1-NRF2 Axis in Renal Cell Carcinoma. Cancers (Basel) 2020; 12:E3458. [PMID: 33233657 PMCID: PMC7699726 DOI: 10.3390/cancers12113458] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/13/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
NRF2 is a transcription factor that coordinates the antioxidant response in many different tissues, ensuring cytoprotection from endogenous and exogenous stress stimuli. In the kidney, its function is essential in appropriate cellular response to oxidative stress, however its aberrant activation supports progression, metastasis, and resistance to therapies in renal cell carcinoma, similarly to what happens in other nonrenal cancers. While at the moment direct inhibitors of NRF2 are not available, understanding the molecular mechanisms that regulate its hyperactivation in specific tumor types is crucial as it may open new therapeutic perspectives. Here, we focus our attention on renal cell carcinoma, describing how NRF2 hyperactivation can contribute to tumor progression and chemoresistance. Furthermore, we highlight the mechanism whereby the many pathways that are generally altered in these tumors converge to dysregulation of the KEAP1-NRF2 axis.
Collapse
Affiliation(s)
| | - Alessandra Boletta
- IRCCS San Raffaele Scientific Institute, Molecular Basis of Cystic Kidney Diseases, Division of Genetics and Cell Biology, 20132 Milan, Italy;
| |
Collapse
|
9
|
Smolková K, Mikó E, Kovács T, Leguina-Ruzzi A, Sipos A, Bai P. Nuclear Factor Erythroid 2-Related Factor 2 in Regulating Cancer Metabolism. Antioxid Redox Signal 2020; 33:966-997. [PMID: 31989830 PMCID: PMC7533893 DOI: 10.1089/ars.2020.8024] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Nuclear factor erythroid 2 (NFE2)-related factor 2 (NFE2L2, or NRF2) is a transcription factor predominantly affecting the expression of antioxidant genes. NRF2 plays a significant role in the control of redox balance, which is crucial in cancer cells. NRF2 activation regulates numerous cancer hallmarks, including metabolism, cancer stem cell characteristics, tumor aggressiveness, invasion, and metastasis formation. We review the molecular characteristics of the NRF2 pathway and discuss its interactions with the cancer hallmarks previously listed. Recent Advances: The noncanonical activation of NRF2 was recently discovered, and members of this pathway are involved in carcinogenesis. Further, cancer-related changes (e.g., metabolic flexibility) that support cancer progression were found to be redox- and NRF2 dependent. Critical Issues: NRF2 undergoes Janus-faced behavior in cancers. The pro- or antineoplastic effects of NRF2 are context dependent and essentially based on the specific molecular characteristics of the cancer in question. Therefore, systematic investigation of NRF2 signaling is necessary to clarify its role in cancer etiology. The biggest challenge in the NRF2 field is to determine which cancers can be targeted for better clinical outcomes. Further, large-scale genomic and transcriptomic studies are missing to correlate the clinical outcome with the activity of the NRF2 system. Future Directions: To exploit NRF2 in a clinical setting in the future, the druggable members of the NRF2 pathway should be identified. In addition, it will be important to study how the modulation of the NRF2 system interferes with cytostatic drugs and their combinations.
Collapse
Affiliation(s)
- Katarína Smolková
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences (IPHYS CAS), Prague, Czech Republic
| | - Edit Mikó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
| | - Tünde Kovács
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Alberto Leguina-Ruzzi
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences (IPHYS CAS), Prague, Czech Republic
| | - Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary.,Faculty of Medicine, Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
10
|
Wang Y, Li C, Li J, Wang G, Li L. Non-Esterified Fatty Acid-Induced Reactive Oxygen Species Mediated Granulosa Cells Apoptosis Is Regulated by Nrf2/p53 Signaling Pathway. Antioxidants (Basel) 2020; 9:antiox9060523. [PMID: 32545880 PMCID: PMC7346109 DOI: 10.3390/antiox9060523] [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: 05/27/2020] [Revised: 06/12/2020] [Accepted: 06/12/2020] [Indexed: 11/17/2022] Open
Abstract
Negative energy balance (NEB) during the perinatal period can affect dairy cow follicular development and reduce the fecundity. Non-esterified fatty acid (NEFA) concentration is elevated during NEB, and is known to be toxic for multiple cell types. In the ovary, NEB increased NEFA, and may influences follicular growth and development. However, the effect and mechanism of NEFA on granulosa cells (GCs) in vitro remains unknown. In this study, we found that NEFA dose-dependently induced apoptosis in primary cultured granulosa cells. Mechanistically, our data showed that NEFA significantly increased reactive oxygen species (ROS) levels, resulting in the activation of endoplasmic reticulum stress (ERS) and eventually cell apoptosis in GCs. Moreover, NEFA also increased the phosphorylation levels of ERK1/2 and p38MAPK pathways, upregulated the expression of p53 and potentially promoted its translocation to the nuclear, thus transcriptionally activated Bax, a downstream gene of this pathway. NEFA also promoted nuclear factor E2 (Nrf2) expression and its level in the nuclear. To elucidate the mechanism of NEFA action, N-acetyl-l-cysteine (NAC), a ROS scavenger was used to verify the role of ROS in NEFA induced apoptosis of GCs. NAC pretreatment reversed the NEFA-induced ERS-related protein and apoptosis-related protein levels. Meanwhile, NAC pretreatment also blocked the phosphorylation of ERK1/2 and p38 induced by NEFA, and the nucleation of Nrf2 and p53, suggesting that ROS plays a crucial role in regulating the NEFA-induced apoptosis of GCs. Together, these findings provide an improved understanding of the mechanisms underlying GCs apoptosis, which could potentially be useful for improving ovarian function.
Collapse
Affiliation(s)
- Yiru Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.W.); (G.W.)
| | - Chengmin Li
- Jiangsu Key Laboratory of Sericutural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China;
| | - Julang Li
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Genlin Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.W.); (G.W.)
| | - Lian Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.W.); (G.W.)
- Correspondence: ; Tel.: +86-25-8439-5045
| |
Collapse
|
11
|
Chen X, Han K, Zhang T, Qi G, Jiang Z, Hu C. Grass carp (Ctenopharyngodon idella) NRF2 alleviates the oxidative stress and enhances cell viability through upregulating the expression of HO-1. FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:417-428. [PMID: 31758371 DOI: 10.1007/s10695-019-00729-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 11/01/2019] [Indexed: 06/10/2023]
Abstract
As a member of the Cap 'n' Collar (CNC) family, NRF2 contains a basic leucine zipper (bZip) and can regulate the downstream target gene heme oxygenase 1 (HO-1) in response to oxidative stress. In the present study, a grass carp (Ctenopharyngodon idella) NRF2 ORF was cloned and identified. The largest ORF (1782 bp) encodes a polypeptide of 593 amino acids. The deduced amino acid sequence of grass carp NRF2 (CiNRF2) contains a well-conserved DNA-binding domain (BRLZ domain). Phylogenetic tree analysis revealed that CiNRF2 has a closer evolutionary relationship with other fish counterparts. After CIK (C. idellus kidney) cells were persistently stimulated with tunicamycin (TM), CiNRF2 was significantly upregulated from 12 to 36 h. Then, the expression was dropped at 48 h post-infection. Additionally, when TM or TG (thapsigargin) stimulated CIK cells, overexpression of CiNRF2 in cells downregulated the expression of Bip mRNA, a marker protein of oxidative stress, suggesting that fish NRF2 can alleviate the oxidative stress level induced by TM or TG. To study the protective mechanism of fish NRF2, the DNA sequences of CiNRF2 and CiATF4 (grass carp ATF4) were separately sub-cloned into the expression vectors pEGFP and pCMV-Flag for co-immunoprecipitation and GST pull-down assays. These assays showed that CiNRF2 can combine with CiATF4 through its Neh1 domain. Meanwhile, we cloned grass carp HO-1 promoter sequence and constructed the recombinant plasmid of pGL3-HO-1. Soon afterwards, pGL3-HO-1 was co-transfected into grass carp ovary (CO) cells with pcDNA3.1-CiNRF2 or pcDNA3.1-CiATF4, respectively. The results showed that the luciferase activity of pGL3-HO-1 in the overexpressed CiNRF2 plus CiATF4 cells was significantly increased, along with the increase of cell viability (~ 133%). However, when HO-1 was knocked down in cells, CiNRF2 was unable to perform its function. These results demonstrated that CiNRF2 was effective in protecting grass carp against the oxidative stress induced by TM and increasing cell viability by upregulating HO-1 expression.
Collapse
Affiliation(s)
- Xin Chen
- Department of Bioscience, College of Life Science, Nanchang University, Nanchang, 330031, China
- Poyang Lake Key Laboratory of Environment and Resource Utilization, Ministry of Education, Nanchang University, Nanchang, China
| | - Kun Han
- Department of Bioscience, College of Life Science, Nanchang University, Nanchang, 330031, China
- Poyang Lake Key Laboratory of Environment and Resource Utilization, Ministry of Education, Nanchang University, Nanchang, China
| | - Tao Zhang
- Department of Bioscience, College of Life Science, Nanchang University, Nanchang, 330031, China
- Poyang Lake Key Laboratory of Environment and Resource Utilization, Ministry of Education, Nanchang University, Nanchang, China
| | - Guoqin Qi
- Department of Bioscience, College of Life Science, Nanchang University, Nanchang, 330031, China
- Poyang Lake Key Laboratory of Environment and Resource Utilization, Ministry of Education, Nanchang University, Nanchang, China
| | - Zeyin Jiang
- Department of Bioscience, College of Life Science, Nanchang University, Nanchang, 330031, China
- Poyang Lake Key Laboratory of Environment and Resource Utilization, Ministry of Education, Nanchang University, Nanchang, China
| | - Chengyu Hu
- Department of Bioscience, College of Life Science, Nanchang University, Nanchang, 330031, China.
- Poyang Lake Key Laboratory of Environment and Resource Utilization, Ministry of Education, Nanchang University, Nanchang, China.
| |
Collapse
|
12
|
Antioxidation and Antiapoptosis Characteristics of Heme Oxygenase-1 Enhance Tumorigenesis of Human Prostate Carcinoma Cells. Transl Oncol 2019; 13:102-112. [PMID: 31810001 PMCID: PMC6909070 DOI: 10.1016/j.tranon.2019.10.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 12/31/2022] Open
Abstract
Heme oxygenase-1 (HO-1) has antiinflammatory and antioxidant properties and is deemed as a tissue protector. However, effects of HO-1 in prostate cancer remain in controversy. We evaluated the role of HO-1 in prostate carcinoma in vitro and in vivo. Overexpression of HO-1 did not affect prostate cell proliferation in the normal condition but enhanced cell proliferation under serum starvation. HO-1 overexpression enhanced cell invasion of PC-3 cells through epithelial–mesenchymal transition (EMT) induction, which was supported by increased Slug, N-cadherin, and vimentin expressions. In the xenograft animal study, HO-1 overexpression enhanced PC-3 cell tumor growth in vivo. HO-1 attenuated reactive oxygen species induced by H2O2 or pyocyanin treatment in PC-3 and DU145 cells. HO-1 further reduced PC-3 and DU145 cell apoptosis induced by H2O2 or serum starvation. Our results suggested that HO-1 was able to increase prostate carcinoma cell invasion in vitro and tumor growth in vivo. The EMT induction and antioxidant and antiapoptotic effects of HO-1 in the prostate carcinoma cells may be responsible for these findings.
Collapse
|
13
|
Potential Applications of NRF2 Inhibitors in Cancer Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8592348. [PMID: 31097977 PMCID: PMC6487091 DOI: 10.1155/2019/8592348] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/10/2019] [Accepted: 02/28/2019] [Indexed: 02/07/2023]
Abstract
The NRF2/KEAP1 pathway represents one of the most important cell defense mechanisms against exogenous or endogenous stressors. Indeed, by increasing the expression of several cytoprotective genes, the transcription factor NRF2 can shelter cells and tissues from multiple sources of damage including xenobiotic, electrophilic, metabolic, and oxidative stress. Importantly, the aberrant activation or accumulation of NRF2, a common event in many tumors, confers a selective advantage to cancer cells and is associated to malignant progression, therapy resistance, and poor prognosis. Hence, in the last years, NRF2 has emerged as a promising target in cancer treatment and many efforts have been made to identify therapeutic strategies aimed at disrupting its prooncogenic role. By summarizing the results from past and recent studies, in this review, we provide an overview concerning the NRF2/KEAP1 pathway, its biological impact in solid and hematologic malignancies, and the molecular mechanisms causing NRF2 hyperactivation in cancer cells. Finally, we also describe some of the most promising therapeutic approaches that have been successfully employed to counteract NRF2 activity in tumors, with a particular emphasis on the development of natural compounds and the adoption of drug repurposing strategies.
Collapse
|
14
|
Liu J, Liu B, Guo Y, Chen Z, Sun W, Gao W, Wu H, Wang Y. Key miRNAs and target genes played roles in the development of clear cell renal cell carcinoma. Cancer Biomark 2019; 23:279-290. [PMID: 30198869 DOI: 10.3233/cbm-181558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Clear cell renal cell carcinoma (CCRCC) is the most aggressive form of renal cell carcinoma (RCC). OBJECTIVE This study was aimed to identify the differentially expressed miRNAs and target genes in CCRCC. METHODS The miRNA and mRNA next-generation sequencing data were downloaded from The Cancer Genome Atlas (TCGA) dataset. Differential expression analysis was performed, followed by correlation analysis of miRNA-mRNA. Functional enrichment and survival analysis was also performed. RESULTS Seven hundred and eighty-seven patients with CCRCC from TCGA data portal were included in this study. A total of 52 differentially expressed miRNAs were identified in CCRCC. Then 2361 differentially expressed genes (DEGs) were identified. Prediction analysis and correlation analysis revealed that 89 miRNA-mRNA pairs were not only predicted by algorithms but also had a significant inverse relationship. Several differentially expressed miRNAs such as hsa-mir-501 and their target genes including AK1, SLC25A15 and PCDHGC3 had a significant prognostic value for CCRCC patients. CONCLUSIONS Alterations of differentially expressed miRNAs and target genes may be involved in the development of CCRCC and can be considered as the prognostic markers for CCRCC.
Collapse
|
15
|
Deng Y, Wu Y, Zhao P, Weng W, Ye M, Sun H, Xu M, Wang C. The Nrf2/HO-1 axis can be a prognostic factor in clear cell renal cell carcinoma. Cancer Manag Res 2019; 11:1221-1230. [PMID: 30799949 PMCID: PMC6369848 DOI: 10.2147/cmar.s188046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE To study the protein expression level of Nrf2/HO-1 in clear cell renal cell carcinoma (ccRCC) and adjacent normal tissue and to explore its relationship with clinicopathological characteristics and prognosis in ccRCC patients. MATERIALS AND METHODS In total, 152 ccRCC patients with available follow-up and clinical data were enrolled, and sample microarrays were prepared for immunohistochemistry studies. The human ccRCC cell lines 786-O, OS-RC-2, A498, and ACHN were cultured for immunofluorescence. The protein concentrations of five ccRCC patients' tumor and adjacent normal renal tissues were prepared for Western blotting. Chi-squared tests, Fisher's exact test, Kaplan-Meier analyses, log-rank tests, and Cox regression were performed for statistical analyses. RESULTS The immunoreactivity results showed that the Nrf2 and HO-1 proteins were found in consistent locations in vitro and were expressed both in ccRCC and adjacent normal tissues. The two proteins were localized in the cytoplasm and nucleus of RCC tumor cells and in adjacent normal tissue cells. The expression levels of Nrf2 and HO-1 were significantly higher in ccRCC tissues than in the adjacent normal tissues. The Nrf2 protein level was found to be significantly correlated with the tumor size. Additionally, higher protein expression levels of Nrf2 and HO-1 were also correlated with worse overall survival outcomes and could potentially be used to predict the prognosis of ccRCC patients. CONCLUSION Our study provides an important theoretical basis for evaluating the clinical prognosis of ccRCC patients, which implies that the Nrf2/HO-1 axis can be a prognostic factor in ccRCC.
Collapse
Affiliation(s)
- Yu Deng
- Department of Pathology, Fudan University Shanghai Cancer Centre, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yong Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Gynaecologic Oncology, Fudan University Shanghai Cancer Centre, Shanghai, China
| | - Ping Zhao
- Department of Pathology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,
| | - Weiwei Weng
- Department of Pathology, Fudan University Shanghai Cancer Centre, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Min Ye
- Department of Pathology, Fudan University Shanghai Cancer Centre, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hui Sun
- Department of Pathology, Fudan University Shanghai Cancer Centre, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Centre, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chaofu Wang
- Department of Pathology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,
| |
Collapse
|
16
|
Activation of c-Met in cancer cells mediates growth-promoting signals against oxidative stress through Nrf2-HO-1. Oncogenesis 2019; 8:7. [PMID: 30647407 PMCID: PMC6333845 DOI: 10.1038/s41389-018-0116-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 12/09/2018] [Accepted: 12/20/2018] [Indexed: 12/28/2022] Open
Abstract
Any imbalance between reactive oxygen species (ROS) generation and the anti-oxidant capacity lead to cellular oxidative stress. Many chemotherapeutic agents mediate their cytotoxic functions through the generation of ROS. c-Met, a receptor tyrosine kinase, is over-expressed in renal cancer and plays very crucial role(s) in its growth and survival. Here, we show that c-Met activation protected renal cancer cells from ROS, oxidative stress and cytotoxicity induced by the anti-cancer agent sorafenib (used for renal cancer treatment); and it markedly attenuated sorafenib-induced DNA damage. Activated c-Met promoted the anti-apoptotic proteins (Bcl-2 and Bcl-xL) and inhibited apoptotic cleaved caspase-3. We found that the cytoprotective function of c-Met against sorafenib-induced ROS generation and apoptosis was mediated primarily through the activation of anti-oxidant Nrf2-HO-1. c-Met promoted the nuclear localization of Nrf2 and hindered its binding with the inhibitory protein Keap1. Silencing of Nrf2 attenuated the protective action of c-Met against sorafenib-induced oxidative stress. To evaluate the physiological significance of our findings, in a tumor xenograft model, we observed that a combination treatment with pharmacological inhibitors of c-Met and it's anti-oxidant downstream effecter HO-1 markedly reduced the growth of renal tumor in vivo; it increased the oxidative stress, DNA damage and apoptotic markers in the tumor xenografts, along with reduced tumor vessel density. Our observations indicate that the c-Met-Nrf2-HO-1 pathway plays a vital role in relieving ROS-mediated oxidative stress of renal tumors. Targeting this pathway can significantly increase the oxidative stress to promote apoptotic death of cancer cells.
Collapse
|
17
|
Metformin Promotes HaCaT Cell Apoptosis through Generation of Reactive Oxygen Species via Raf-1-ERK1/2-Nrf2 Inactivation. Inflammation 2018; 41:948-958. [PMID: 29549478 DOI: 10.1007/s10753-018-0749-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Although metformin (MET) may be useful for the treatment of psoriasis, the mechanisms underlying its method of action have yet to be fully elucidated. Here, the relationship between MET function and reactive oxygen species (ROS) levels and the underlying mechanism were explored in human immortalized keratinocyte cell line (HaCaT). HaCaT cells were incubated with MET at 0, 10, 20, 40, and 60 mM for 24 h. The cell viability was evaluated by the CCK-8 assay. The cell apoptosis rate and intracellular ROS levels were examined using flow cytometry. The protein expression and the phosphorylation levels of nuclear factor erythroid-derived 2 related factor 2 (Nrf2), Raf-1, and ERK1/2 were assessed by Western blot. The specific ROS scavenger N-acetyl-cysteine (NAC) and the specific Nrf2 agonist Oltipraz (OPZ) were used to analyze the effect of MET. MET decreased HaCaT cell proliferation and induced HaCaT cell apoptosis in a dose-dependent manner. MET was found to elevate intracellular ROS levels in a dose-dependent manner, while pretreatment with NAC attenuated these effects. MET inhibits the protein expression and the phosphorylation levels of Nrf2. The combination of OPZ and MET can significantly increase the cell viability, decrease the rate of apoptosis, and attenuate the intracellular ROS levels relative to MET alone. MET inhibits the protein expression and the phosphorylation levels of Raf-1 and ERK1/2. MET was found to attenuate Raf-1-ERK1/2 signaling in HaCaT cells to suppress the expression and phosphorylation levels of Nrf2, which contributed to the intracellular generation of ROS and the pro-apoptotic effects of MET.
Collapse
|
18
|
Saahene RO, Wang J, Wang ML, Agbo E, Pang D. The Antitumor Mechanism of Paeonol on CXCL4/CXCR3-B Signals in Breast Cancer Through Induction of Tumor Cell Apoptosis. Cancer Biother Radiopharm 2018; 33:233-240. [PMID: 29847158 DOI: 10.1089/cbr.2018.2450] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Paeonol, a phenolic component from the root bark of Paeonia moutan, has been identified to possess antitumor effects. However, the effect of paeonol and the mechanism of CXCL4/CXCR3-B signals in paeonol-induced breast cancer cell remain unknown. MATERIALS AND METHODS After MDA-MB-231 cells were pretreated with paeonol or DMSO, the proliferation activity was detected by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide), Hoechst, Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and Annexin-V/propidium iodide staining flow cytometry. Western blot and immunohistochemistry of human breast cancer and noncancerous tissues were performed to determine the molecular alteration of CXCL4/CXCR3-B signals. RESULTS Compared with the control, paeonol-treated breast cancer cells had low proliferation activity and high apoptotic index, indicating that paeonol induces breast cancer cell apoptosis. Western blot and immunohistochemistry showed that paeonol increased CXCR3-B signal, downregulated CXCL4, heme oxygenase (HO-1) with a corresponding increased BACH1, and decreased nuclear factor E2-related factor 2 (Nrf2). CONCLUSIONS Thus, CXCL4/CXCR3-B may be involved in the mechanism of apoptosis induced by paeonol in breast cancer cells by regulating the expression of BACH1 and Nrf2 to downregulating HO-1 and promote apoptosis. Therefore, the authors suggest paeonol has a significant growth inhibitory effect on breast cancer cells, which may be related to the induction of apoptosis.
Collapse
Affiliation(s)
- Roland O Saahene
- 1 Department of Immunology, College of Basic Medicine, Jiamusi University , People's Republic of China
| | - Jianjie Wang
- 1 Department of Immunology, College of Basic Medicine, Jiamusi University , People's Republic of China
| | - Mo-Lin Wang
- 1 Department of Immunology, College of Basic Medicine, Jiamusi University , People's Republic of China
| | - Elvis Agbo
- 2 Department of Anatomy, Jiamusi University , People's Republic of China
| | - Dezhi Pang
- 1 Department of Immunology, College of Basic Medicine, Jiamusi University , People's Republic of China
| |
Collapse
|
19
|
Park SA, Lee MH, Na HK, Surh YJ. 4-Hydroxyestradiol induces mammary epithelial cell transformation through Nrf2-mediated heme oxygenase-1 overexpression. Oncotarget 2018; 8:164-178. [PMID: 27438141 PMCID: PMC5352084 DOI: 10.18632/oncotarget.10516] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/12/2016] [Indexed: 12/30/2022] Open
Abstract
Estrogen (17β-estradiol, E2) undergoes oxidative metabolism by CYP1B1 to form 4-hydroxyestradiol (4-OHE2), a putative carcinogenic metabolite of estrogen. Our previous study showed that 4-OHE2-induced production of reactive oxygen species contributed to neoplastic transformation of human breast epithelial (MCF-10A) cells. In this study, 4-OHE2, but not E2, increased the expression of heme oxygenase-1 (HO-1), a sensor and regulator of oxidative stress, in MCF-10A cells. Silencing the HO-1 gene in MCF-10A cells suppressed 4-OHE2-induced cell proliferation and transformation. In addition, subcutaneous administration of 4-OHE2 markedly enhanced the growth of the MDA-MB-231 human breast cancer xenografts, which was retarded by zinc protoporphyrin, a pharmacological inhibitor of HO-1. 4-OHE2-induced HO-1 expression was mediated by NF-E2-related factor 2 (Nrf2). We speculate that an electrophilic quinone formed as a consequence of oxidation of 4-OHE2 binds directly to Kelch-like ECH-associated protein 1 (Keap1), an inhibitory protein that sequesters Nrf2 in the cytoplasm. This will diminish association between Nrf2 and Keap1. 4-OHE2 failed to interrupt the interaction between Keap1 and Nrf2 and to induce HO-1 expression in Keap1-C273S or C288S mutant cells. Lano-LC-ESI-MS/MS analysis in MCF-10A-Keap1-WT cells which were treated with 4-OHE2 revealed that the peptide fragment containing Cys288 gained a molecular mass of 287.15 Da, equivalent to the addition of a single molecule of 4-OHE2-derived ortho-quinones.
Collapse
Affiliation(s)
- Sin-Aye Park
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, South Korea
| | - Mee-Hyun Lee
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, South Korea
| | - Hye-Kyung Na
- Department of Food and Nutrition, College of Human Ecology, Sungshin Women's University, Seoul 136-742, South Korea
| | - Young-Joon Surh
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, South Korea.,Cancer Research Institute, Seoul National University, Seoul 110-799, South Korea
| |
Collapse
|
20
|
Molecular Pathogenesis of Liver Injury in Hereditary Tyrosinemia 1. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 959:49-64. [PMID: 28755183 DOI: 10.1007/978-3-319-55780-9_4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Untreated HT1 rapidly degenerates into very severe liver complications often resulting in liver cancer. The molecular basis of the pathogenic process in HT1 is still unclear. The murine model of FAH-deficiency is a suitable animal model, which represents all phenotypic and biochemical manifestations of the human disease on an accelerated time scale. After removal of the drug 2-(2-N-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione (NTBC), numerous signaling pathways involved in cell proliferation, differentiation and cancer are rapidly deregulated in FAH deficient mice. Among these, the Endoplasmic reticulum (ER) pathway, the heat stress response (HSR), the Nrf2, MEK and ERK pathways, are highly represented. The p21 and mTOR pathways critical regulators of proliferation and tumorigenesis have also been found to be dysregulated. The changes in these pathways are described and related to the development of liver cancer.
Collapse
|
21
|
Honokiol inhibits c-Met-HO-1 tumor-promoting pathway and its cross-talk with calcineurin inhibitor-mediated renal cancer growth. Sci Rep 2017; 7:5900. [PMID: 28724911 PMCID: PMC5517643 DOI: 10.1038/s41598-017-05455-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/13/2017] [Indexed: 02/06/2023] Open
Abstract
Honokiol (HNK) is a small molecule with potent anti-inflammatory and anti-tumorigenic properties; yet the molecular targets of HNK are not well studied. Hyperactivation of the receptor tyrosine kinase c-Met and overexpression of the cytoprotective enzyme heme oxygenase-1 (HO-1) play a critical role in the growth and progression of renal cell carcinoma (RCC). Interestingly, the calcineurin inhibitor (CNI) cyclosporine A (CsA), an immunosuppressant used to prevent allograft rejection, can also increase the risk of RCC in transplant patients. We studied the potential role of c-Met signaling axis on CNI-induced renal tumor growth and tested the anti-tumor efficacy of HNK. Importantly, CNI treatment promoted c-Met induction and enhanced c-Met-induced Ras activation. We found that HNK treatment effectively down-regulated both c-Met phosphorylation and Ras activation in renal cancer cells. It inhibited the expression of both c-Met- and CNI-induced HO-1, and promoted cancer cell apoptosis. In vivo, HNK markedly inhibited CNI-induced renal tumor growth; and it decreased the expression of phospho-c-Met and HO-1 and reduced blood vessel density in tumor tissues. Our results suggest a novel mechanism(s) by which HNK exerts its anti-tumor activity through the inhibition of c-Met-Ras-HO-1 axis; and it can have significant therapeutic potential to prevent post-transplantation cancer in immunosuppressed patients.
Collapse
|
22
|
Shi Y, An D, Liu Y, Feng Q, Fang X, Pan G, Wang Q. Propoxur enhances MMP-2 expression and the corresponding invasion of human breast cancer cells via the ERK/Nrf2 signaling pathway. Oncotarget 2017; 8:87107-87123. [PMID: 29152067 PMCID: PMC5675619 DOI: 10.18632/oncotarget.19081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 06/20/2017] [Indexed: 11/25/2022] Open
Abstract
Propoxur is considered a prime etiological suspect of increasing tumor incidence, but the role is still undefined. In this study, two human breast cancer cells lines, MCF-7 and MDA-MB-231 cells, were used as cell models. Cells were respectively treated with 0, 0.01, 1, or 100 μM propoxur. PD98059, a MEK inhibitor, was administered to block the ERK/MAPK pathway. Migration and reactive oxygen species were measured by wound healing and Transwell assays, and flow cytometry. Protein expression and subcellular location were detected by western blotting and immunofluorescence staining, respectively. Results showed that propoxur treatment enhanced cell migration and invasion in a dose-dependent manner, while MMP-2 expression, but not MMP-9, was significantly increased in two cell lines. Meanwhile, the treatment increased intracellular reactive oxygen species, Nrf2 expression and nuclear translocation, and ERK1/2 phosphorylation. Inversely, inhibition of ERK1/2 activation with PD98059 significantly attenuated propoxur-induced Nrf2 expression and nuclear translocation. Moreover, PD98059 suppressed propoxur-induced cell migration and invasion, and MMP-2 overexpression. Collectively, these results indicate that propoxur can trigger reactive oxygen species overproduction, further promoting breast cancer cell migration and invasion by regulating the ERK/Nrf2 signaling pathways.
Collapse
Affiliation(s)
- Yunxiang Shi
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing 100071, China
| | - Daizhi An
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing 100071, China
| | - Yiping Liu
- Beijing Municipal Public Security Hospital, Beijing Municipal Public Security Bureau, Beijing 100006, China
| | - Qiong Feng
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing 100071, China
| | - Xu Fang
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing 100071, China
| | - Guilan Pan
- Department of Physiology, BaoTou Medical College, Inner Mongolia University of Science and Technology, Baotou 014040, China
| | - Qiang Wang
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing 100071, China
| |
Collapse
|
23
|
Noh SJ, Kim KM, Jang KY. Individual and co-expression patterns of nerve growth factor and heme oxygenase-1 predict shorter survival of gastric carcinoma patients. Diagn Pathol 2017; 12:48. [PMID: 28679437 PMCID: PMC5498870 DOI: 10.1186/s13000-017-0644-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 06/30/2017] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Nerve growth factor (NGF) is a neurotrophic factor which regulates cell development and proliferation. Recently, it has been suggested that NGF induces heme oxygenase-1 (HO1) expression, and that both NGF and HO1 are involved in the progression of malignant human tumors. However, exact roles of NGF and HO1 in tumorigenesis remain controversial. Therefore, we investigated the expression and correlation of NGF and HO1 in human gastric carcinoma tissues. METHODS We examined immunohistochemical expression of NGF and HO1 in 167 gastric carcinomas and compared with various prognostic clinicopathological factors. RESULTS The expression of NGF and HO1 was positive in 40% (67/167) and 51% (85/167) of cases, respectively, and their expression was significantly correlated with each other (p < 0.001). Individual expression patterns of NGF and HO1, and co-expression pattern of these two molecules were significantly associated with shorter survival by univariate analysis. HO1 expression (overall survival; p < 0.001, relapse-free survival; p = 0.002) and co-expression pattern of NGF and HO1 (overall survival; p = 0.002, relapse-free survival; p = 0.003) were independent poor prognostic indicators of gastric carcinoma patients by multivariate analysis. CONCLUSIONS These results demonstrate that the individual and co-expression patterns of NGF and HO1 might be used as prognostic indicators for gastric carcinoma patients.
Collapse
Affiliation(s)
- Sang Jae Noh
- Department of Forensic Medicine, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, 20, Geonji-ro, Deokjin-gu, Jeonju, Jeonbuk, 54907, Republic of Korea
| | - Kyoung Min Kim
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, 20, Geonji-ro, Deokjin-gu, Jeonju, Jeonbuk, 54907, Republic of Korea
| | - Kyu Yun Jang
- Department of Pathology, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital and Research Institute for Endocrine Sciences, 20, Geonji-ro, Deokjin-gu, Jeonju, Jeonbuk, 54907, Republic of Korea.
| |
Collapse
|
24
|
|
25
|
Peng CY, You BJ, Lee CL, Wu YC, Lin WH, Lu TL, Chang FC, Lee HZ. The Roles of 4β-Hydroxywithanolide E from Physalis peruviana on the Nrf2-Anti-Oxidant System and the Cell Cycle in Breast Cancer Cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2016; 44:617-36. [PMID: 27109152 DOI: 10.1142/s0192415x16500348] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
4[Formula: see text]-Hydroxywithanolide E is an active component of the extract of Physalis peruviana that has been reported to exhibit antitumor effects. Although the involvement of reactive oxygen species (ROS) production and the ataxia-telangiectasia mutated protein (ATM)-dependent DNA damage signaling pathway in 4[Formula: see text]-hydroxywithanolide E-induced apoptosis of breast cancer MCF-7 cells was demonstrated in our previous study, the relationship between ROS production and the cellular defense system response in 4[Formula: see text]-hydroxywithanolide E-induced cell death requires further verification. The present study suggests that ROS play an important role in 4[Formula: see text]-hydroxywithanolide E-induced MCF-7 cell death in which anti-oxidants, such as glutathione or N-acetylcysteine, can resist the 4[Formula: see text]-hydroxywithanolide E-induced accumulation of ROS and cell death. Furthermore, N-acetylcysteine or glutathione can reverse the 4[Formula: see text]-hydroxywithanolide E-induced changes in the cell cycle distribution and the expression of cell cycle regulators. We found that the 4[Formula: see text]-hydroxywithanolide E-induced ROS accumulation was correlated with the upregulation of Nrf2 and Nrf2-downstream genes, such as antioxidative defense enzymes. In general, the activity of Nrf2 is regulated by the Ras signalling pathway. However, we demonstrated that Nrf2 was activated during 4[Formula: see text]-hydroxywithanolide E-induced MCF-7 cell death in spite of the 4[Formula: see text]-hydroxywithanolide E-induced inhibition of the Ras/Raf/ERK pathway. The activity and protein expression of superoxide dismutase and catalase were involved in the 4[Formula: see text]-hydroxywithanolide E-induced ROS production in MCF-7 cells. Furthermore, 4[Formula: see text]-hydroxywithanolide E was demonstrated to significantly reduce the sizes of the tumor nodules in the human breast cancer MDA-MB231 xenograft tumor model.
Collapse
Affiliation(s)
- Chieh Yu Peng
- School of Pharmacy, China Medical University Hospital, Taichung, Taiwan
| | - Bang Jau You
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, Taiwan
| | - Chia Lin Lee
- Department of Cosmeceutics and Graduate Institute of Cosmeceutics, China Medical University, Taichung, Taiwan
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
| | - Yang Chang Wu
- School of Pharmacy, China Medical University Hospital, Taichung, Taiwan
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung, Taiwan
| | - Wen Hsin Lin
- School of Pharmacy, China Medical University Hospital, Taichung, Taiwan
| | - Te Ling Lu
- School of Pharmacy, China Medical University Hospital, Taichung, Taiwan
| | - Fei-Ching Chang
- Pharmacy Department, Tainan Municipal An-Nan Hospital-China Medical University, Tainan, Taiwan
| | - Hong Zin Lee
- School of Pharmacy, China Medical University Hospital, Taichung, Taiwan
- Pharmacy Department, Tainan Municipal An-Nan Hospital-China Medical University, Tainan, Taiwan
| |
Collapse
|
26
|
Genrich G, Kruppa M, Lenk L, Helm O, Broich A, Freitag-Wolf S, Röcken C, Sipos B, Schäfer H, Sebens S. The anti-oxidative transcription factor Nuclear factor E2 related factor-2 (Nrf2) counteracts TGF-β1 mediated growth inhibition of pancreatic ductal epithelial cells -Nrf2 as determinant of pro-tumorigenic functions of TGF-β1. BMC Cancer 2016; 16:155. [PMID: 26915435 PMCID: PMC4766703 DOI: 10.1186/s12885-016-2191-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 02/17/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Nuclear factor E2 related factor-2 (Nrf2) is an oxidative stress inducible transcription factor being essential in regulating cell homeostasis. Thus, acute induction of Nrf2 in epithelial cells exposed to inflammation confers protection from oxidative cell damage and mutagenesis supporting an anti-tumorigenic role for Nrf2. However, pancreatic ductal adenocarcinoma (PDAC) is characterized by persistent Nrf2 activity conferring therapy resistance which points to a pro-tumorigenic role of Nrf2. A similar dichotomous role in tumorigenesis is described for the Transforming Growth Factor-beta 1 (TGF-β1). The present study therefore aimed at elucidating whether the switch of Nrf2 function towards a tumor promoting one relates to the modulation of TGF-β1 induced cell responses and whether this might occur early in PDAC development. METHODS In situ analysis comprised immunohistochemical stainings of activated (phosphorylated) Nrf2 and Ki67 in pancreatic tissues containing normal ducts and pancreatic intraepithelial neoplasia (PanINs). In vitro, Nrf2 levels in benign (H6c7-pBp), premalignant (H6c7-kras) and malignant (Colo357) pancreatic ductal epithelial cells were modulated by Nrf2 specific siRNA or Nrf2 overexpression. Then, the effect of Nrf2 alone and in combination with TGF-β1 on cell growth and survival was investigated by cell counting, Ki67 staining and apoptosis assays. The underlying cell signaling was investigated by western blotting. Statistical analysis was performed by Shapiro-Wilk test for normal distribution. Parametric data were analyzed by one-way ANOVA, while non-parametric data were analyzed by Kruskal-Wallis one-way ANOVA on ranks. RESULTS Significantly elevated expression of activated Nrf2 and Ki67 could be detected in PanINs but not in normal pancreatic ductal epithelium. While the effect of Nrf2 on basal cell growth of H6c7-pBp, H6c7-kras and Colo357 cells was minor, it clearly attenuated the growth inhibiting effects of TGF-β1 in all cell lines. This enhanced Nrf2-mediated cell survival was predominantly based on an enhanced proliferative activity. Accordingly, expression of p21 expression along with expression of phospho-p38 and phospho-Smad3 was diminished whereas Erk-phosphorylation was enhanced under these conditions. CONCLUSIONS Overall, our data demonstrate that Nrf2 being elevated in early precursor lesions counteracts the growth inhibiting function of TGF-β1 already in benign and premalignant pancreatic ductal epithelial cells. This could represent one fundamental mechanism underlying the functional switch of both- TGF-β1 and Nrf2 - which may manifest already in early stages of PDAC development.
Collapse
Affiliation(s)
- Geeske Genrich
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| | - Marcus Kruppa
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| | - Lennart Lenk
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| | - Ole Helm
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| | - Anna Broich
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| | - Sandra Freitag-Wolf
- Institute of Medical Informatics and Statistics, UKSH Campus Kiel, Brunswiker Str. 10, 24105, Kiel, Germany.
| | - Christoph Röcken
- Department of Pathology, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 14, 24105, Kiel, Germany.
| | - Bence Sipos
- Department of Pathology and Neuropathology, University Hospital Tübingen, Liebermeisterstr. 8, 72076, Tübingen, Germany.
| | - Heiner Schäfer
- Laboratory of Molecular Gastroenterology & Hepatology, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 6, 24105, Kiel, Germany.
| | - Susanne Sebens
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| |
Collapse
|
27
|
Liao JC, Lee KT, You BJ, Lee CL, Chang WT, Wu YC, Lee HZ. Raf/ERK/Nrf2 signaling pathway and MMP-7 expression involvement in the trigonelline-mediated inhibition of hepatocarcinoma cell migration. Food Nutr Res 2015; 59:29884. [PMID: 26699938 PMCID: PMC4689951 DOI: 10.3402/fnr.v59.29884] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 11/07/2015] [Accepted: 11/07/2015] [Indexed: 01/05/2023] Open
Abstract
Background Trigonelline occurs in many dietary food plants and has been found to have anti-carcinogenic activity. Trigonelline is also found in coffee which is one of the most widely consumed beverages. Many epidemiological studies have reported that coffee consumption has an inverse relationship with the risk of cirrhosis or hepatocellular carcinoma. It would be interesting to investigate whether trigonelline is an ideal chemoprevent agent to prevent cancer progression. Methods The protein expression was performed by western blotting. The trigonelline content in snow pea (Pisum sativum) was analyzed by high-performance liquid chromatography (HPLC). The migratory activity of human hepatocarcinoma cells (Hep3B) was assessed by using a wound migration assay. The percentage of each phase in the cell cycle was analyzed on a FACScan flow cytometer. Gene expression was detected by real-time reverse transcriptase-polymerase chain reaction techniques. Native gel analysis was performed to analyze the activity of superoxide dismutase (SOD), catalase and glutathione peroxidase. Results According to the data of HPLC analysis, P. sativum, which is a popular vegetable, has relatively high content of trigonelline. Our findings suggest that trigonelline is an efficient compound for inhibiting Hep3B cell migration. Trigonelline inhibited the migration of hepatoma cells at concentrations of 75–100 µM without affecting proliferation. Raf/ERK/Nrf2 protein levels and further downstream antioxidative enzymes activity, such as SOD, catalase, and glutathione peroxidase, significantly decreased after treatment with 100 µM of trigonelline for 24 h. The migration inhibition of trigonelline is also related to its ability to regulate the matrix metalloproteinases 7 (MMP-7) gene expression. Conclusions In this study, protein kinase Cα (PKCα) and Raf/ERK/Nrf2 signaling pathway and MMP-7 gene expression were involved in the trigonelline-mediated migration inhibition of Hep3B cells. We also demonstrated that trigonelline inhibits Hep3B cell migration through downregulation of nuclear factor E2-related factor 2–dependent antioxidant enzymes activity. This study analyzed the trigonelline content in a popular vegetable, snow pea, as a representative proof to prove that trigonelline is often found in the daily intake of food. Our finding suggested that trigonelline should be a useful chemopreventive agent derived from the daily intake of food to prevent cancer progression.
Collapse
Affiliation(s)
- Jung Chun Liao
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Kun Tsung Lee
- Department of Oral Hygiene, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Bang Jau You
- School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Chia Lin Lee
- Department of Cosmeceutics and Graduate Institute of Cosmeceutics, China Medical University, Taichung, Taiwan
| | - Wen Te Chang
- School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Yang Chang Wu
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Hong-Zin Lee
- School of Pharmacy, China Medical University, Taichung, Taiwan.,Pharmacy Department, Tainan Municipal An-Nan Hospital-China Medical University, Tainan, Taiwan;
| |
Collapse
|
28
|
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: 221] [Impact Index Per Article: 22.1] [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
|
29
|
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: 142] [Impact Index Per Article: 14.2] [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
|
30
|
Angileri F, Roy V, Morrow G, Scoazec JY, Gadot N, Orejuela D, Tanguay RM. Molecular changes associated with chronic liver damage and neoplastic lesions in a murine model of hereditary tyrosinemia type 1. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2603-17. [PMID: 26360553 DOI: 10.1016/j.bbadis.2015.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 08/28/2015] [Accepted: 09/04/2015] [Indexed: 01/06/2023]
Abstract
Hereditary tyrosinemia type 1 (HT1) is the most severe inherited metabolic disease of the tyrosine catabolic pathway, with a progressive hepatic and renal injury and a fatal outcome if untreated. Toxic metabolites accumulating in HT1 have been shown to elicit endoplasmic reticulum (ER) stress response, and to induce chromosomal instability, cell cycle arrest and apoptosis perturbation. Although many studies have concentrated on elucidating these events, the molecular pathways responsible for development of hepatocellular carcinoma (HCC) still remain unclear. In this study the fah knockout murine model (fah(-/-)) was used to investigate the cellular signaling implicated in the pathogenesis of HT1. Fah(-/-) mice were subjected to drug therapy discontinuation (Nitisinone withdrawal), and livers were analyzed at different stages of the disease. Monitoring of mice revealed an increasing degeneration of the overall physiological conditions following drug withdrawal. Histological analysis unveiled diffuse hepatocellular damage, steatosis, oval-like cells proliferation and development of liver cell adenomas. Immunoblotting results revealed a progressive and chronic activation of stress pathways related to cell survival and proliferation, including several stress regulators such as Nrf2, eIF2α, CHOP, HO-1, and some members of the MAPK signaling cascade. Impairment of stress defensive mechanisms was also shown by microarray analysis in fah(-/-) mice following prolonged therapy interruption. These results suggest that a sustained activation of stress pathways in the chronic HT1 progression might play a central role in exacerbating liver degeneration.
Collapse
Affiliation(s)
- Francesca Angileri
- Laboratoire de génétique cellulaire et développementale,IBIS et PROTEO,Département de Biologie Moléculaire,Biochimie Médicale et Pathologie,Faculté de Médecine,1030 Ave de la médecine,Université Laval,Québec G1V 0A6,Canada
| | - Vincent Roy
- Laboratoire de génétique cellulaire et développementale,IBIS et PROTEO,Département de Biologie Moléculaire,Biochimie Médicale et Pathologie,Faculté de Médecine,1030 Ave de la médecine,Université Laval,Québec G1V 0A6,Canada
| | - Geneviève Morrow
- Laboratoire de génétique cellulaire et développementale,IBIS et PROTEO,Département de Biologie Moléculaire,Biochimie Médicale et Pathologie,Faculté de Médecine,1030 Ave de la médecine,Université Laval,Québec G1V 0A6,Canada
| | - Jean Yves Scoazec
- Service Central d'anatomie et de Cytologie Pathologiques,Hôpital Edouard-Herriot,69437 Lyon Cedex 03,France
| | - Nicolas Gadot
- Service Central d'anatomie et de Cytologie Pathologiques,Hôpital Edouard-Herriot,69437 Lyon Cedex 03,France
| | - Diana Orejuela
- Laboratoire de génétique cellulaire et développementale,IBIS et PROTEO,Département de Biologie Moléculaire,Biochimie Médicale et Pathologie,Faculté de Médecine,1030 Ave de la médecine,Université Laval,Québec G1V 0A6,Canada
| | - Robert M Tanguay
- Laboratoire de génétique cellulaire et développementale,IBIS et PROTEO,Département de Biologie Moléculaire,Biochimie Médicale et Pathologie,Faculté de Médecine,1030 Ave de la médecine,Université Laval,Québec G1V 0A6,Canada.
| |
Collapse
|
31
|
Zheng WX, Yan F, Xue Q, Wu GJ, Qin WJ, Wang FL, Qin J, Tian CJ, Yuan JL. Heme oxygenase-1 is a predictive biomarker for therapeutic targeting of advanced clear cell renal cell carcinoma treated with sorafenib or sunitinib. Onco Targets Ther 2015; 8:2081-8. [PMID: 26309414 PMCID: PMC4539079 DOI: 10.2147/ott.s86222] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background We analyzed the expression of heme oxygenase-1 (HO-1) in patients undergoing radical nephrectomy for advanced clear cell renal cell carcinoma (CC-RCC) and evaluated the effects of the targeted therapies treated with sorafenib and sunitinib. Methods Expression of HO-1 in cancer tissue from 66 patients was measured by immunohis-tochemical staining. The patients received either oral sorafenib (n=40) or oral sunitinib (n=26) within 4 weeks after nephrectomy and were followed up long term to determine the tumor response and prognosis. Results Our current study revealed a high HO-1 expression level in 57.6% (38/66) of patients and a low HO-1 expression level in 42.4% (28/66) of patients with CC-RCC. The study also revealed that patients with high HO-1 expression did not have a higher objective response rate (2.6% versus 53.6%, P<0.01), clinical benefit rate (47.4% versus 92.9%, P<0.01), longer progression-free survival (4.4 versus 42 months, P=0.022), or overall survival (χ2=4.775, P=0.029) than patients with low HO-1 expression. In the low HO-1 level group, a higher tumor response rate and a longer survival time was achieved in patients who received sorafenib or sunitinib. Multivariate analysis showed that HO-1 expression was an independent prognostic factor for tumor response and overall survival. Conclusion High expression of HO-1 was associated with a lower tumor response rate and a shorter overall survival time when compared with low expression of HO-1. Overall, HO-1 expression might be a useful biomarker for predicting the response to sunitinib and sorafenib for patients with metastatic CC-RCC.
Collapse
Affiliation(s)
- Wan-Xiang Zheng
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Fei Yan
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Qin Xue
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Guo-Jun Wu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Wei-Jun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Fu-Li Wang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Jun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Chun-Juan Tian
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Jian-Lin Yuan
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
32
|
|
33
|
Balan M, Mier y Teran E, Waaga-Gasser AM, Gasser M, Choueiri TK, Freeman G, Pal S. Novel roles of c-Met in the survival of renal cancer cells through the regulation of HO-1 and PD-L1 expression. J Biol Chem 2015; 290:8110-20. [PMID: 25645920 DOI: 10.1074/jbc.m114.612689] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The receptor tyrosine kinase c-Met is overexpressed in renal cancer cells and can play major role in the growth and survival of tumor. We investigated how the c-Met-mediated signaling through binding to its ligand hepatocyte growth factor (HGF) can modulate the apoptosis and immune escape mechanism(s) of renal cancer cells by the regulations of novel molecules heme oxygenase-1 (HO-1) and programmed death-1 ligand 1 (PD-L1). We found that HGF/c-Met-mediated signaling activated the Ras/Raf pathway and down-regulated cancer cell apoptosis; and it was associated with the overexpression of cytoprotective HO-1 and anti-apoptotic Bcl-2/Bcl-xL. c-Met-induced HO-1 overexpression was regulated at the transcriptional level. Next, we observed that c-Met induction markedly up-regulated the expression of the negative co-stimulatory molecule PD-L1, and this can be prevented following treatment of the cells with pharmacological inhibitors of c-Met. Interestingly, HGF/c-Met-mediated signaling could not induce PD-L1 at the optimum level when either Ras or HO-1 was knocked down. To study the functional significance of c-Met-induced PD-L1 expression, we performed a co-culture assay using mouse splenocytes (expressing PD-L1 receptor PD-1) and murine renal cancer cells (RENCA, expressing high PD-L1). We observed that the splenocyte-mediated apoptosis of cancer cells during co-culture was markedly increased in the presence of either c-Met inhibitor or PD-L1 neutralizing antibody. Finally, we found that both c-Met and PD-L1 are significantly up-regulated and co-localized in human renal cancer tissues. Together, our study suggests a novel mechanism(s) by which c-Met can promote increased survival of renal cancer cells through the regulation of HO-1 and PD-L1.
Collapse
Affiliation(s)
- Murugabaskar Balan
- From the Division of Nephrology, Boston Children's Hospital, Harvard Medical School, and
| | | | - Ana Maria Waaga-Gasser
- Department of Surgery, Molecular Oncology, and Immunology, University of Wurzburg, 97080 Wurzburg, Germany
| | - Martin Gasser
- Department of Surgery, Molecular Oncology, and Immunology, University of Wurzburg, 97080 Wurzburg, Germany
| | - Toni K Choueiri
- Harvard Medical School, and Dana Farber Cancer Institute, Boston, Massachusetts 02115 and
| | - Gordon Freeman
- Harvard Medical School, and Dana Farber Cancer Institute, Boston, Massachusetts 02115 and
| | - Soumitro Pal
- From the Division of Nephrology, Boston Children's Hospital, Harvard Medical School, and
| |
Collapse
|
34
|
Geismann C, Arlt A, Sebens S, Schäfer H. Cytoprotection "gone astray": Nrf2 and its role in cancer. Onco Targets Ther 2014; 7:1497-518. [PMID: 25210464 PMCID: PMC4155833 DOI: 10.2147/ott.s36624] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Nrf2 has gained great attention with respect to its pivotal role in cell and tissue protection. Primarily defending cells against metabolic, xenobiotic and oxidative stress, Nrf2 is essential for maintaining tissue integrity. Owing to these functions, Nrf2 is regarded as a promising drug target in the chemoprevention of diseases, including cancer. However, much evidence has accumulated that the beneficial role of Nrf2 in cancer prevention essentially depends on the tight control of its activity. In fact, the deregulation of Nrf2 is a critical determinant in oncogenesis and found in many types of cancer. Therefore, amplified Nrf2 activity has profound effects on the phenotype of tumor cells, including radio/chemoresistance, apoptosis protection, invasiveness, antisenescence, autophagy deficiency, and angiogenicity. The deregulation of Nrf2 can result from various epigenetic and genetic alterations directly affecting Nrf2 control or from the complex interplay of Nrf2 with numerous oncogenic signaling pathways. Additionally, alterations of the cellular environment, eg, during inflammation, contribute to Nrf2 deregulation and its persistent activation. Therefore, the status of Nrf2 as anti- or protumorigenic is defined by many different modalities. A better understanding of these modalities is essential for the safe use of Nrf2 as an activation target for chemoprevention on the one hand and as an inhibition target in cancer therapy on the other. The present review mainly addresses the conditions that promote the oncogenic function of Nrf2 and the resulting consequences providing the rationale for using Nrf2 as a target structure in cancer therapy.
Collapse
Affiliation(s)
- Claudia Geismann
- Laboratory of Molecular Gastroenterology, Department of Internal Medicine I, Universitätsklinikum Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Alexander Arlt
- Laboratory of Molecular Gastroenterology, Department of Internal Medicine I, Universitätsklinikum Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Susanne Sebens
- Inflammatory Carcinogenesis Research Group, Institute of Experimental Medicine, Universitätsklinikum Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Heiner Schäfer
- Laboratory of Molecular Gastroenterology, Department of Internal Medicine I, Universitätsklinikum Schleswig-Holstein Campus Kiel, Kiel, Germany
| |
Collapse
|
35
|
Understanding life and death decisions in human leukaemias. Biochem Soc Trans 2014; 42:747-51. [PMID: 25109952 DOI: 10.1042/bst20140127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Human leukaemia cells have an often unique ability to either undergo apoptotic cell death mechanisms or, at other times, undergo proliferative expansion, sometimes to the same stimulus such as the pluripotent cytokine TNFα (tumour necrosis factor α). This potential for life/death switching helps us to understand the molecular signalling machinery that underlies these cellular processes. Furthermore, looking at the involvement of these switching signalling pathways that may be aberrant in leukaemia informs us of their importance in cancer tumorigenesis and how they may be targeted pharmacologically to treat various types of human leukaemias. Furthermore, these important pathways may play a crucial role in acquired chemotherapy resistance and should be studied further to overcome in the clinic many drug-resistant forms of blood cancers. In the present article, we uncover the relationship that exists in human leukaemia life/death switching between the anti-apoptotic pro-inflammatory transcription factor NF-κB (nuclear factor κB) and the cytoprotective antioxidant-responsive transcription factor Nrf2 (nuclear factor-erythroid 2-related factor 2). We also discuss recent findings that reveal a major role for Btk (Bruton's tyrosine kinase) in both lymphocytic and myeloid forms of human leukaemias and lymphomas.
Collapse
|
36
|
Qi D, Ouyang C, Wang Y, Zhang S, Ma X, Song Y, Yu H, Tang J, Fu W, Sheng L, Yang L, Wang M, Zhang W, Miao L, Li T, Huang X, Dong H. HO-1 attenuates hippocampal neurons injury via the activation of BDNF–TrkB–PI3K/Akt signaling pathway in stroke. Brain Res 2014; 1577:69-76. [DOI: 10.1016/j.brainres.2014.06.031] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 06/24/2014] [Accepted: 06/25/2014] [Indexed: 01/07/2023]
|
37
|
Abstract
PURPOSE OF REVIEW Heme oxygenase activity, possessed by an inducible heme oxygenase-1 (HO-1) and a constitutive isoform (HO-2), catalyzes the conversion of heme to biliverdin, liberates iron, and generates carbon monoxide. First shown in acute kidney injury (AKI), HO-1 is now recognized as a protectant against diverse insults in assorted tissues. This review summarizes recent contributions to the field of HO-1 and AKI. RECENT FINDINGS Recent findings elucidate the following: the transcriptional regulation and significance of human HO-1 in AKI; the protective effects of HO-1 in age-dependent and sepsis-related AKI, cardiorenal syndromes, and acute vascular rejection in renal xenografts; the role of heme oxygenase in tubuloglomerular feedback and renal resistance to injury; the basis for cytoprotection by HO-1; the protective properties of ferritin and carbon monoxide; HO-1 and the AKI-chronic kidney disease transition; HO-1 as a biomarker in AKI; the role of HO-1 in mediating the protective effects of specific cytokines, stem cells, and therapeutic agents in AKI; and HO-2 as a protectant in AKI. SUMMARY Recent contributions support, and elucidate the basis for, the induction of HO-1 as a protectant against AKI. Translating such therapeutic potential into a therapeutic reality requires well tolerated and effective modalities for upregulating HO-1 and/or administering its products, which, optimally, should be salutary even when AKI is already established.
Collapse
|
38
|
Liu YS, Li HS, Qi DF, Zhang J, Jiang XC, Shi K, Zhang XJ, Zhang XH. Zinc protoporphyrin IX enhances chemotherapeutic response of hepatoma cells to cisplatin. World J Gastroenterol 2014; 20:8572-8582. [PMID: 25024611 PMCID: PMC4093706 DOI: 10.3748/wjg.v20.i26.8572] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 01/16/2014] [Accepted: 04/03/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of zinc protoporphyrin IX on the response of hepatoma cells to cisplatin and the possible mechanism involved.
METHODS: Cytotoxicity was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Apoptosis was determined by a flow cytometric assay. Western blotting was used to measure protein expression. Heme oxygenase (HO)-1 activity was measured by determining the level of bilirubin generated in isolated microsomes. Reactive oxygen species (ROS) production was monitored by flow cytometry. Caspase-3 activity was measured with a colorimetric assay kit. Mice were inoculated with 1 × 107 tumor cells subcutaneously into the right flanks. All mice were sacrificed 6 wk after the first treatment and tumors were weighed and measured.
RESULTS: Overexpression of HO-1 in HepG2 cell line was associated with increased chemoresistance to cis-diaminedichloroplatinum (cisplatin; CDDP) compared to other cell lines in vitro. Inhibition of HO-1 expression or activity by zinc protoporphyrin IX (ZnPP IX) markedly augmented CDDP-mediated cytotoxicity towards all liver cancer cell lines in vitro and in vivo. In contrast, induction of HO-1 with hemin increased resistance of tumor cells to CDDP-mediated cytotoxicity in vitro and in vivo. Furthermore, cells treated with ZnPP IX plus CDDP exhibited marked production of intracellular ROS and caspase-3 activity, which paralleled the incidence of cell apoptosis, whereas hemin decreased cellular ROS and caspase-3 activity induced by CDDP.
CONCLUSION: ZnPP IX increases cellular sensitivity and susceptibility of liver cancer cell lines to CDDP and this may represent a mechanism of increasing ROS.
Collapse
|
39
|
Wang X, Guo X, Yu W, Li C, Gui Y, Cai Z. Expression of methionine adenosyltransferase 2A in renal cell carcinomas and potential mechanism for kidney carcinogenesis. BMC Cancer 2014; 14:196. [PMID: 24636201 PMCID: PMC4003826 DOI: 10.1186/1471-2407-14-196] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 03/06/2014] [Indexed: 12/16/2022] Open
Abstract
Background Methionine adenosyltransferase 2A (MAT2A) is an enzyme that catalyzes the formation of S-adenosylmethionine (SAMe) by joining methionine and ATP. SAMe is a methyl donor for transmethylation and has an important role for DNA and/or protein methylation. MAT2A is expressed widely in many tissues especially in kidney. Several studies have demonstrated that there are abnormal expressions of MAT2A in several kinds of cancers such as liver and colon cancers. But the relationship of MAT2A between renal cell carcinomas (RCC) is less understood. Methods The mRNA expression level of the MAT2A gene was determined in 24 RCC patients and 4 RCC cell lines, using real-time quantitative-polymerase chain reaction (RT-PCR). The MAT2A protein content was measured by western blotting and immunohistochemical analysis in 55 RCC patients. The mRNA levels of heme oxygenase-1 (HO-1) and cyclooxygenase-2 (COX-2) were also analysized in patients using RT-PCR. The correlations between the MAT2A and HO-1 as well as COX-2 were analyzed with nonparametric Spearman method. Results MAT2A transcript was significantly downregulated in cancer tissues compared to normal tissues (P < 0.05). Immunohistochemical analysis and western blotting indicated that level of MAT2A protein was decreased in cancer tissues. The statistical analysis reveals a negative correlation between MAT2A and HO-1 expression in RCC patients and cell lines (P < 0.01). Conclusions This study demonstrated that MAT2A was lower expression in cancer tissues, suggesting that it may be involved in the development of RCC. MAT2A is a transcriptional corepressor for HO-1 expression by supplying SAM for methyltransferases, which may be one of potential mechanism of MAT2A as tumor suppressor in kidney carcinogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhiming Cai
- Shenzhen Key Laboratory of Genitourinary Tumor, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, Guangdong, China.
| |
Collapse
|
40
|
Na HK, Surh YJ. Oncogenic potential of Nrf2 and its principal target protein heme oxygenase-1. Free Radic Biol Med 2014; 67:353-65. [PMID: 24200599 DOI: 10.1016/j.freeradbiomed.2013.10.819] [Citation(s) in RCA: 355] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 10/28/2013] [Accepted: 10/29/2013] [Indexed: 10/26/2022]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is an essential component of cellular defense against a vast variety of endogenous and exogenous insults, including oxidative stress. Nrf2 acts as a master switch in the circuits upregulating the expression of various stress-response proteins, especially heme oxygenase-1 (HO-1). Paradoxically, however, recent studies have demonstrated oncogenic functions of Nrf2 and its major target protein HO-1. Levels of Nrf2 and HO-1 are elevated in many different types of human malignancies, which may facilitate the remodeling of the tumor microenvironment making it advantageous for the autonomic growth of cancer cells, metastasis, angiogenesis, and tolerance to chemotherapeutic agents and radiation and photodynamic therapy. In this context, the cellular stress response or cytoprotective signaling mediated via the Nrf2-HO-1 axis is hijacked by cancer cells for their growth advantage and survival of anticancer treatment. Therefore, Nrf2 and HO-1 may represent potential therapeutic targets in the management of cancer. This review highlights the roles of Nrf2 and HO-1 in proliferation of cancer cells, their tolerance/resistance to anticancer treatments, and metastasis or angiogenesis in tumor progression.
Collapse
Affiliation(s)
- Hye-Kyung Na
- Department of Food & Nutrition, College of Human Ecology, Sungshin Women's University, Seoul 142-732, South Korea
| | - Young-Joon Surh
- Tumor Microenvironment Global Core Research Institute, College of Pharmacy, Seoul National University, Seoul 151-742, South Korea; Department of Molecular Medicine and Biopharmaceutical Science, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 151-742, South Korea; Cancer Research Institute, Seoul National University, Seoul 110-744, South Korea.
| |
Collapse
|
41
|
Chung S, Yoon HE, Kim SJ, Kim SJ, Koh ES, Hong YA, Park CW, Chang YS, Shin SJ. Oleanolic acid attenuates renal fibrosis in mice with unilateral ureteral obstruction via facilitating nuclear translocation of Nrf2. Nutr Metab (Lond) 2014; 11:2. [PMID: 24393202 PMCID: PMC3896758 DOI: 10.1186/1743-7075-11-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 01/01/2014] [Indexed: 12/12/2022] Open
Abstract
Background Renal interstitial fibrosis is a common final pathological process in the progression of kidney disease. This is primarily due to oxidative stress, which contributes to renal inflammation and fibrosis. Nuclear factor-erythroid-2-related factor 2 (Nrf2) is known to coordinate induction of genes that encode antioxidant enzymes. We investigated the effects of oleanolic acid, a known Nrf2 activator, on oxidative stress-induced renal inflammation and fibrosis. Methods One day before unilateral ureteral obstruction (UUO) performed in C57BL/6 mice, oleanolic acid treatment was initiated and was continued until 3 and 7 days after UUO. Renal inflammation and fibrosis, markers of oxidative stress, and changes in Nrf2 expression were subsequently evaluated. Results In the obstructed kidneys of UUO mice, oleanolic acid significantly attenuated UUO-induced collagen deposition and fibrosis on day 7. Additionally, significantly less inflammatory cell infiltration, a lower ratio of Bax to Bcl-2 expression, and fewer apoptotic cells on TUNEL staining were observed in the obstructed kidneys of oleanolic acid-treated mice. Oleanolic acid increased the expression of nuclear Nrf2, heme oxygenase-1, NAD(P)H:quinone oxidoreductase 1 and heat shock protein 70, and decreased lipid peroxidation in the obstructed kidney of UUO mice. There were no changes in the expression of total Nrf2 and Kelch-like ECH-associated protein 1, indicating that oleanolic acid enhanced nuclear translocation of Nrf2. Conclusions These results suggest that oleanolic acid may exert beneficial effects on renal fibrosis by increasing nuclear translocation of Nrf2 and subsequently reducing renal oxidative stress.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Seok Joon Shin
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seoul 137-701, Republic of Korea.
| |
Collapse
|
42
|
Balan M, Pal S. A novel CXCR3-B chemokine receptor-induced growth-inhibitory signal in cancer cells is mediated through the regulation of Bach-1 protein and Nrf2 protein nuclear translocation. J Biol Chem 2013; 289:3126-37. [PMID: 24366869 DOI: 10.1074/jbc.m113.508044] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Chemokines and their receptors play diverse roles in regulating cancer growth and progression. The receptor CXCR3 can have two splice variants with opposite functions. CXCR3-A promotes cell growth, whereas CXCR3-B mediates growth-inhibitory signals. However, the negative signals through CXCR3-B in cancer cells are not well characterized. In this study, we found that CXCR3-B-mediated signaling in MCF-7 and T47D breast cancer cells induced apoptotic cell death. Signals through CXCR3-B decreased the levels of the antiapoptotic proteins Bcl-2 and Bcl-xL and increased the expression of apoptotic cleaved poly(ADP-ribose) polymerase. Along with up-regulation in apoptosis, CXCR3-B signals were associated with a decrease in cellular autophagy with reduced levels of the autophagic markers Beclin-1 and LC3B. Notably, CXCR3-B down-regulated the expression of the cytoprotective and antiapoptotic molecule heme oxygenase-1 (HO-1) at the transcriptional level. There was an increased nuclear localization of Bach-1 and nuclear export of Nrf2, which are important negative and positive transcription factors, respectively, for HO-1 expression. We also observed that CXCR3-B promoted the activation of p38 MAPK and the inhibition of ERK-1/2. CXCR3-B could not induce cancer cell apoptosis at the optimal level when we either inhibited p38 activity or knocked down Bach-1. Further, CXCR3-B-induced apoptosis was down-regulated when we overexpressed HO-1. Together, our data suggest that CXCR3-B mediates a growth-inhibitory signal in breast cancer cells through the modulations of nuclear translocation of Bach-1 and Nrf2 and down-regulation of HO-1. We suggest that the induction of CXCR3-B-mediated signaling can serve as a novel therapeutic approach where the goal is to promote tumor cell apoptosis.
Collapse
Affiliation(s)
- Murugabaskar Balan
- From the Division of Nephrology, Boston Children's Hospital, Boston, Massachusetts 02115 and
| | | |
Collapse
|
43
|
Banerjee P, Basu A, Arbiser JL, Pal S. The natural product honokiol inhibits calcineurin inhibitor-induced and Ras-mediated tumor promoting pathways. Cancer Lett 2013; 338:292-9. [PMID: 23752066 DOI: 10.1016/j.canlet.2013.05.036] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 05/14/2013] [Accepted: 05/27/2013] [Indexed: 11/19/2022]
Abstract
Although calcineurin inhibitors (CNIs) are very useful in preventing allograft rejection, they can mediate a rapid progression of post-transplantation malignancies. The CNI cyclosporine A (CsA) can promote renal tumor growth through activation of the proto-oncogene ras and over-expression of the angiogenic cytokine VEGF; the ras activation also induces over-expression of the cytoprotective enzyme HO-1, which promotes survival of renal cancer cells. Here, we show that the natural product honokiol significantly inhibited CsA-induced and Ras-mediated survival of renal cancer cells through the down-regulations of VEGF and HO-1. Thus, honokiol treatment may help to prevent tumor-promoting effects of CsA in transplant patients.
Collapse
Affiliation(s)
- Pallavi Banerjee
- Division of Nephrology, Children's Hospital, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
44
|
Cong ZX, Wang HD, Wang JW, Zhou Y, Pan H, Zhang DD, Zhu L. ERK and PI3K signaling cascades induce Nrf2 activation and regulate cell viability partly through Nrf2 in human glioblastoma cells. Oncol Rep 2013; 30:715-22. [PMID: 23708697 DOI: 10.3892/or.2013.2485] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 04/19/2013] [Indexed: 11/06/2022] Open
Abstract
The ERK and PI3K signaling cascades are aberrantly activated in human glioblastoma cells, resulting in the dysregulation of numerous downstream transcription factors. The dark side of the transcription factor, NF-E2-related factor 2 (Nrf2) in human cancer has been revealed. It has been accepted that high levels of Nrf2 promote tumor progression. In the present study, we investigated the effect of the ERK and PI3K signaling cascades on Nrf2 in human glioblastoma cells. Immunohistochemical staining for Nrf2 in clinical specimens showed that the expression and nuclear localization of Nrf2 were increased in human glioblastoma tissues when compared to peritumoral normal tissues. In addition, we detected decreased nuclear localization of Nrf2 following combined treatment with ERK and PI3K inhibitors in three human glioblastoma cell lines and selected the cell line (U251) most sensitive to the inhibitors for further study. Our data demonstrated that inhibition of ERK and PI3K not only suppressed the nuclear accumulation of Nrf2 protein but also decreased the expression of the Nrf2 protein. In addition, combined inhibition of ERK and PI3K also decreased the mRNA levels of Nrf2 target genes. Finally, we found that Nrf2 overexpression partly reversed the ERK and PI3K inhibitor-induced inhibition of cell viability. Therefore, the ERK and PI3K signaling cascades regulate the expression and activation of Nrf2 and control cell viability partly through Nrf2 in U251 human glioblastoma cells. Thus, targeting the ERK and PI3K signaling cascades for Nrf2 activation may provide new methods for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Zi-Xiang Cong
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, PR China
| | | | | | | | | | | | | |
Collapse
|
45
|
Do MT, Kim HG, Khanal T, Choi JH, Kim DH, Jeong TC, Jeong HG. Metformin inhibits heme oxygenase-1 expression in cancer cells through inactivation of Raf-ERK-Nrf2 signaling and AMPK-independent pathways. Toxicol Appl Pharmacol 2013; 271:229-38. [PMID: 23707609 DOI: 10.1016/j.taap.2013.05.010] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 04/29/2013] [Accepted: 05/10/2013] [Indexed: 01/12/2023]
Abstract
Resistance to therapy is the major obstacle to more effective cancer treatment. Heme oxygenase-1 (HO-1) is often highly up-regulated in tumor tissues, and its expression is further increased in response to therapies. It has been suggested that inhibition of HO-1 expression is a potential therapeutic approach to sensitize tumors to chemotherapy and radiotherapy. In this study, we tested the hypothesis that the anti-tumor effects of metformin are mediated by suppression of HO-1 expression in cancer cells. Our results indicate that metformin strongly suppresses HO-1 mRNA and protein expression in human hepatic carcinoma HepG2, cervical cancer HeLa, and non-small-cell lung cancer A549 cells. Metformin also markedly reduced Nrf2 mRNA and protein levels in whole cell lysates and suppressed tert-butylhydroquinone (tBHQ)-induced Nrf2 protein stability and antioxidant response element (ARE)-luciferase activity in HepG2 cells. We also found that metformin regulation of Nrf2 expression is mediated by a Keap1-independent mechanism and that metformin significantly attenuated Raf-ERK signaling to suppress Nrf2 expression in cancer cells. Inhibition of Raf-ERK signaling by PD98059 decreased Nrf2 mRNA expression in HepG2 cells, confirming that the inhibition of Nrf2 expression is mediated by an attenuation of Raf-ERK signaling in cancer cells. The inactivation of AMPK by siRNA, DN-AMPK or the pharmacological AMPK inhibitor compound C, revealed that metformin reduced HO-1 expression in an AMPK-independent manner. These results highlight the Raf-ERK-Nrf2 axis as a new molecular target in anticancer therapy in response to metformin treatment.
Collapse
Affiliation(s)
- Minh Truong Do
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon, South Korea
| | | | | | | | | | | | | |
Collapse
|
46
|
Bauckman KA, Haller E, Flores I, Nanjundan M. Iron modulates cell survival in a Ras- and MAPK-dependent manner in ovarian cells. Cell Death Dis 2013; 4:e592. [PMID: 23598404 PMCID: PMC3668627 DOI: 10.1038/cddis.2013.87] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 02/01/2013] [Accepted: 02/19/2013] [Indexed: 02/07/2023]
Abstract
Ovarian cancer is a leading cause of cancer death in women in the United States. While the majority of ovarian cancers are serous, some rarer subtypes (i.e. clear cell) are often associated with endometriosis, a benign gynecological disease. Iron is rich in the cyst fluid of endometriosis-associated ovarian cancers and induces persistent oxidative stress. The role of iron, an essential nutrient involved in multiple cellular functions, in normal ovarian cell survival and ovarian cancer remains unclear. Iron, presented as ferric ammonium citrate (FAC), dramatically inhibits cell survival in ovarian cancer cell types associated with Ras mutations, while it is without effect in immortalized normal ovarian surface epithelial (T80) and endometriotic epithelial cells (lacking Ras mutations). Interestingly, FAC induced changes in cytoplasmic vacuolation concurrently with increases in LC3-II levels (an autophagy marker); these changes occurred in an ATG5/ATG7-dependent, beclin-1/hVps34-independent, and Ras-independent manner. Knockdown of autophagy mediators in HEY ovarian cancer cells reversed FAC-induced LC3-II levels, but there was little effect on reversing the cell death response. Intriguingly, transmission electron microscopy of FAC-treated T80 cells demonstrated abundant lysosomes (confirmed using Lysotracker) rich in iron particles, which occurred in a Ras-independent manner. Although the mitogen-activated protein kinase (MAPK) inhibitor, U0126, reversed FAC-induced LC3-II/autophagic punctae and lysosomes in a Ras-independent manner, it was remarkable that U0126 reversed cell death in malignant ovarian cells associated with Ras mutations. Moreover, FAC increased heme oxygenase-1 expression in H-Ras-overexpressing T80 cells, which was associated with increased cell death when overexpressed in T80 cells. Disruption of intracellular iron levels, via chelation of intracellular iron (deferoxamine), was also detrimental to malignant ovarian cell survival; thus, homeostatic intracellular iron levels are essential for cell survival. Collectively, our results implicate iron in modulating cell death in a Ras- and MAPK-dependent manner in ovarian cancer cells.
Collapse
Affiliation(s)
- K A Bauckman
- Moffitt Cancer Center and Research Institute, Cancer Biology Program, Tampa, FL, USA
| | - E Haller
- Department of Integrative Biology, University of South Florida, Tampa, FL, USA
| | - I Flores
- Departments of Microbiology, Obstetrics and Gynecology, Ponce School of Medicine and Health Sciences, Ponce, Puerto Rico
| | - M Nanjundan
- Moffitt Cancer Center and Research Institute, Cancer Biology Program, Tampa, FL, USA
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL, USA
| |
Collapse
|
47
|
Hu J, He Y, Yan M, Zhu C, Ye W, Zhu H, Chen W, Zhang C, Zhang Z. Dose dependent activation of retinoic acid-inducible gene-I promotes both proliferation and apoptosis signals in human head and neck squamous cell carcinoma. PLoS One 2013; 8:e58273. [PMID: 23484008 PMCID: PMC3587586 DOI: 10.1371/journal.pone.0058273] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 01/31/2013] [Indexed: 12/25/2022] Open
Abstract
The retinoic-acid-inducible gene (RIG)-like receptor (RLR) family proteins are major pathogen reorganization receptors (PRR) responsible for detection of viral RNA, which initiates antiviral response. Here, we evaluated the functional role of one RLR family member, RIG-I, in human head and neck squamous cell carcinoma (HNSCC). RIG-I is abundantly expressed both in poorly-differentiated primary cancer and lymph node metastasis, but not in normal adjacent tissues. Activation of RIG-I by transfection with low dose of 5'-triphosphate RNA (3p-RNA) induces low levels of interferon and proinflammatory cytokines and promotes NF-κB- and Akt-dependent cell proliferation, migration and invasion. In contrast, activation of RIG-I by a high dose of 3p-RNA induces robust mitochondria-derived apoptosis accompanied by decreased activation of Akt, which is independent of the interferon and TNFα receptor, but can be rescued by over-expression of constitutively active Akt. Furthermore, co-immunoprecipitation experiments indicate that the CARD domain of RIG-I is essential for inducing apoptosis by interacting with caspase-9. Together, our results reveal a dual role of RIG-I in HNSCC through regulating activation of Akt, in which RIG-I activation by low-dose viral dsRNA increases host cell survival, whereas higher level of RIG-I activation leads to apoptosis. These findings highlight the therapeutic potential of dsRNA mediated RIG-I activation in the treatment of HNSCC.
Collapse
Affiliation(s)
- Jingzhou Hu
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue He
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Yan
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Zhu
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weimin Ye
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hanguang Zhu
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wantao Chen
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenping Zhang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyuan Zhang
- Department of Oral & Maxillofacial-Head & Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
48
|
Armeni T, Ercolani L, Urbanelli L, Magini A, Magherini F, Pugnaloni A, Piva F, Modesti A, Emiliani C, Principato G. Cellular redox imbalance and changes of protein S-glutathionylation patterns are associated with senescence induced by oncogenic H-ras. PLoS One 2012; 7:e52151. [PMID: 23284910 PMCID: PMC3527427 DOI: 10.1371/journal.pone.0052151] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 11/09/2012] [Indexed: 12/31/2022] Open
Abstract
H-Ras oncogene requires deregulation of additional oncogenes or inactivation of tumor suppressor proteins to increase cell proliferation rate and transform cells. In fact, the expression of the constitutively activated H-RasV12 induces cell growth arrest and premature senescence, which act like barriers in pre-neoplastic lesions. In our experimental model, human fibroblasts transfected with H-RasV12 show a dramatic modification of morphology. H-RasV12 expressing cells also show premature senescence followed by cell death, induced by autophagy and apoptosis. In this context, we provide evidence that in H-RasV12 expressing cells, the premature senescence is associated with cellular redox imbalance as well as with altered post-translation protein modification. In particular, redox imbalance is due to a strong reduction of total antioxidant capacity, and significant decrease of glutathione level. As the reversible addition of glutathione to cysteinyl residues of proteins is an important post-translational regulative modification, we investigated S-glutathionylation in cells expressing active H-Ras. In this contest we observed different S-glutathionylation patterns in control and H-RasV12 expressing cells. Particularly, the GAPDH enzyme showed S-glutathionylation increase and significant enzyme activity depletion in H-Ras V12 cells. In conclusion, we proposed that antioxidant defense reduction, glutathione depletion and subsequent modification of S-glutathionylation of target proteins contribute to arrest cell growth, leading to death of fibroblasts expressing constitutively active H-Ras oncogene, thus acting as oncogenic barriers that obstacle the progression of cell transformation.
Collapse
Affiliation(s)
- Tatiana Armeni
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica delle Marche, Ancona, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Banerjee P, Basu A, Wegiel B, Otterbein LE, Mizumura K, Gasser M, Waaga-Gasser AM, Choi AM, Pal S. Heme oxygenase-1 promotes survival of renal cancer cells through modulation of apoptosis- and autophagy-regulating molecules. J Biol Chem 2012; 287:32113-23. [PMID: 22843690 DOI: 10.1074/jbc.m112.393140] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The cytoprotective enzyme heme oxygenase-1 (HO-1) is often overexpressed in different types of cancers and promotes cancer progression. We have recently shown that the Ras-Raf-ERK pathway induces HO-1 to promote survival of renal cancer cells. Here, we examined the possible mechanisms underlying HO-1-mediated cell survival. Considering the growing evidence about the significance of apoptosis and autophagy in cancer, we tried to investigate how HO-1 controls these events to regulate survival of cancer cells. Rapamycin (RAPA) and sorafenib, two commonly used drugs for renal cancer treatment, were found to induce HO-1 expression in renal cancer cells Caki-1 and 786-O; and the apoptotic effect of these drugs was markedly enhanced upon HO-1 knockdown. Overexpression of HO-1 protected the cells from RAPA- and sorafenib-induced apoptosis and also averted drug-mediated inhibition of cell proliferation. HO-1 induced the expression of anti-apoptotic Bcl-xL and decreased the expression of autophagic proteins Beclin-1 and LC3B-II; while knockdown of HO-1 down-regulated Bcl-xL and markedly increased LC3B-II. Moreover, HO-1 promoted the association of Beclin-1 with Bcl-xL and Rubicon, a novel negative regulator of autophagy. Drug-induced dissociation of Beclin-1 from Rubicon and the induction of autophagy were also inhibited by HO-1. Together, our data signify that HO-1 is up-regulated in renal cancer cells as a survival strategy against chemotherapeutic drugs and promotes growth of tumor cells by inhibiting both apoptosis and autophagy. Thus, application of chemotherapeutic drugs along with HO-1 inhibitor may elevate therapeutic efficiency by reducing the cytoprotective effects of HO-1 and by simultaneous induction of both apoptosis and autophagy.
Collapse
Affiliation(s)
- Pallavi Banerjee
- Division of Nephrology, Children's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Lin AH, Chen HW, Liu CT, Tsai CW, Lii CK. Activation of Nrf2 is required for up-regulation of the π class of glutathione S-transferase in rat primary hepatocytes with L-methionine starvation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:6537-6545. [PMID: 22676582 DOI: 10.1021/jf301567m] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Numerous genes expression is regulated in response to amino acid shortage, which helps organisms adapt to amino acid limitation. The expression of the π class of glutathione (GSH) S-transferase (GSTP), a highly inducible phase II detoxification enzyme, is regulated mainly by activates activating protein 1 (AP-1) binding to the enhancer I of GSTP (GPEI). Here we show the critical role of nuclear factor erythroid-2-related factor 2 (Nrf2) in up-regulating GSTP gene transcription. Primary rat hepatocytes were cultured in a methionine-restricted medium, and immunoblotting and RT-PCR analyses showed that methionine restriction time-dependently increased GSTP protein and mRNA expression over a 48 h period. Nrf2 translocation to the nucleus, nuclear proteins binding to GPEI, and antioxidant response element (ARE) luciferase reporter activity were increased by methionine restriction as well as by l-buthionine sulfoximine (BSO), a GSH synthesis inhibitor. Transfection with Nrf2 siRNA knocked down Nrf2 expression and reversed the methionine-induced GSTP expression and GPEI binding activity. Chromatin immunoprecipitation assay confirmed the binding of Nrf2 to the GPEI. Phosphorylation of extracellular signal-regulated kinase 2 (ERK2) was increased in methionine-restricted and BSO-treated cells. ERK2 siRNA abolished methionine restriction-induced Nrf2 nuclear translocation, GPEI binding activity, ARE-luciferase reporter activity, and GSTP expression. Our results suggest that the up-regulation of GSTP gene transcription in response to methionine restriction likely occurs via the ERK-Nrf2-GPEI signaling pathway.
Collapse
Affiliation(s)
- Ai-Hsuan Lin
- School of Nutrition, Chung Shan Medical University, Taichung 402, Taiwan
| | | | | | | | | |
Collapse
|